WO2006121522A2 - Implantable sensors and pumps, anti-scarring agents, and therapeutic compositions - Google Patents

Implantable sensors and pumps, anti-scarring agents, and therapeutic compositions Download PDF

Info

Publication number
WO2006121522A2
WO2006121522A2 PCT/US2006/011726 US2006011726W WO2006121522A2 WO 2006121522 A2 WO2006121522 A2 WO 2006121522A2 US 2006011726 W US2006011726 W US 2006011726W WO 2006121522 A2 WO2006121522 A2 WO 2006121522A2
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
agent
sensor
antagonist
composition
Prior art date
Application number
PCT/US2006/011726
Other languages
French (fr)
Other versions
WO2006121522A3 (en
Inventor
William L. Hunter
Philip M. Toleikis
David M. Gravett
Arpita Maiti
Richard T. Liggins
Aniko Takacs-Cox
Rui Avelar
Pierre E. Signore
Troy A. E. Loss
Anne Hutchinson
Gaye Mcdonald-Jones
Fara Lakhani
Original Assignee
Angiotech International Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Angiotech International Ag filed Critical Angiotech International Ag
Publication of WO2006121522A2 publication Critical patent/WO2006121522A2/en
Publication of WO2006121522A3 publication Critical patent/WO2006121522A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/14Infusion devices, e.g. infusing by gravity; Blood infusion; Accessories therefor
    • A61M5/142Pressure infusion, e.g. using pumps
    • A61M5/14244Pressure infusion, e.g. using pumps adapted to be carried by the patient, e.g. portable on the body
    • A61M5/14276Pressure infusion, e.g. using pumps adapted to be carried by the patient, e.g. portable on the body specially adapted for implantation

Definitions

  • the present invention relates generally to implantable sensors, drug-delivery devices and drug-delivery pump, and more specifically, to compositions and methods for preparing and using such devices to make them resistant to overgrowth by inflammatory and fibrous scar tissue.
  • Implantable drug delivery devices and pumps are a means to provide prolonged, site-specific release of a therapeutic agent for the management of a variety of medical conditions.
  • Drug delivery implants and pumps are generally utilized when a localized pharmaceutical impact is desired (i.e., the condition affects only a specific region) or when systemic delivery of the agent is inefficient or ineffective and leads toxicity, severe side effects, inactivation of the drug prior to reaching the target tissue, poor symptom/disease control, and/or addiction to the medication.
  • Implantable pumps can also deliver systemic drug levels in a constant, regulated manner for extended periods and help patients avoid the "peaks and valleys" of blood-level drug concentrations associated with intermittent systemic dosing.
  • Innumerable drug delivery devices, implants and pumps have been developed for an array of specific medical conditions and the particular construction and delivery mechanism of the device depends on the particular treatment.
  • drug delivery implants and pumps have been used in a variety of clinical applications, including programmable insulin pumps for the treatment of diabetes, intrathecal (in the spine) pumps to administer narcotics (e.g., morphine, fentanyl) for the relief of pain (e.g., cancer, back problems, HIV, post-surgery), local and systemic delivery of chemotherapy for the treatment of cancer (e.g., hepatic artery 5-FU infusion for liver tumors), medications for the treatment of cardiac conditions (e.g., antiarrhythmic drugs for cardiac rhythm abnormalities), intrathecal delivery of anti- spasmotic drugs (e.g., baclofen) for spasticity in neurological disorders (e.g., Multiple Sclerosis, spinal cord injuries, brain injury, cerebral palsy), or local/regional antibiotics for infection management (e.g., osteomyelitis, septic arthritis).
  • narcotics e.g., morphine, fentanyl
  • chemotherapy e.g., cancer, back problems
  • most drug delivery pumps are implanted subcutaneously (under the skin in an easy to access, but discrete location) and consist of a pump unit with a drug reservoir and a flexible catheter through which the drug is delivered to the target tissue.
  • the pump stores and releases prescribed amounts of medication via the catheter to achieve therapeutic drug levels either locally or systemically (depending upon the application).
  • the center of the pump has a self-sealing access port covered by a septum such that a needle can be inserted percutaneously (through both the skin and the septum) to refill the pump with medication as required.
  • Constant-rate pumps are usually powered by gas and are designed to dispense drugs under pressure as a continual dosage at a preprogrammed, constant rate.
  • Programmable-rate pumps utilize a battery-powered pump and a constant pressure reservoir to deliver drugs on a periodic basis in a manner that can be programmed by the physician or the patient.
  • the drug may be delivered in small, discrete doses based on a programmed regimen which can be altered according to an individual's clinical response.
  • Programmable drug delivery pumps may be in communication with an external transmitter which programs the prescribed dosing regimen, including the rate, time and amount of each dose, via low-frequency waves that are transmitted through the skin.
  • Programmable-rate pumps are more widely used and provide superior dosimetry, but because of their complexity, they require more maintenance and have a shorter lifespan.
  • an implantable drug delivery device or pump depends upon the device, particularly the catheter, being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum) and not becoming encapsulated or obstructed by scar tissue.
  • target tissue e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum
  • scar tissue e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum
  • Scarring i.e., fibrosis
  • Scarring can also result from trauma to the anatomical structures and tissue surrounding the implant during implantation of the device.
  • fibrous encapsulation of the device can occur even after a successful implantation if the device is manipulated (some patients continuously "fiddle" with a subcutaneous implant) or irritated by the daily activities of the patient.
  • the catheter tip or lumen may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely.
  • the catheter can become encapsulated by scar (i.e., the body "walls off' the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement from the catheter to the tissues on the other side of the capsule).
  • a device that is frequently (but not always) used in association with a drug delivery pump is an implantable sensor device.
  • An implantable sensor is a device used to detect changes in body function and/or levels of key physiological metabolites, chemistry, hormones or biological factors.
  • Implantable sensors may be used to sense a variety of physical and/or physiological properties, including, but not limited to, optical, mechanical, chemical, electrochemical, temperature, strain, pressure, magnetism, acceleration, ionizing radiation, acoustic wave or chemical changes.
  • sensor technology is combined with implantable drug delivery pumps such that the sensor receives a signal and then, in turn, uses this information to modulate the release kinetics of a drug.
  • implantable pancreas which can continuously detect blood glucose levels (through an implanted sensor) and provide feedback to an implantable pump to modulate the administration of insulin to a diabetic patient.
  • implantable sensors include, blood/tissue glucose monitors, electrolyte sensors, blood constituent sensors, temperature sensors, pH sensors, optical sensors, amperometric sensors, pressure sensors, biosensors, sensing transponders, strain sensors, activity sensors and magnetoresistive sensors.
  • Scarring around the implanted device may degrade the electrical components and characteristics of the device-tissue interface, and the device may fail to function properly. For example, when a "foreign body” response occurs and the implanted sensor becomes encapsulated by scar (Ae., the body "walls off' the sensor with fibrous tissue), the sensor receives inaccurate biological information. If the sensor is detecting conditions inside the capsule, and these conditions are not consistent with those outside the capsule (which is frequently the case), it will produce inaccurate readings. Similarly if the scar tissue alters the flow of physical or chemical information to the detection mechanism of the sensor, the information it processes will not be reflective of those present in the target tissue.
  • scar tissue alters the flow of physical or chemical information to the detection mechanism of the sensor, the information it processes will not be reflective of those present in the target tissue.
  • the present invention discloses pharmaceutical agents which inhibit one or more aspects of the production of excessive fibrous (scar) tissue.
  • the present invention provides compositions for delivery of selected therapeutic agents via medical devices or implants containing sensors or drug delivery pumps, as well as methods for making and using these implants and devices.
  • Compositions and methods are described for coating sensors or pumps with drug-delivery compositions such that the pharmaceutical agent is delivered in therapeutic levels over a period sufficient to prevent the drug delivery catheter and/or the implanted sensor from being encapsulated in fibrous tissue to improve and/or prolong device function.
  • compositions e.g., topicals, injectables, liquids, gels, sprays, microspheres, pastes, wafers
  • an inhibitor of fibrosis e.g., topicals, injectables, liquids, gels, sprays, microspheres, pastes, wafers
  • an inhibitor of fibrosis e.g., an inhibitor of fibrosis
  • numerous specific implantable pumps, sensors and combined devices are described that produce superior clinical results as a result of being coated with agents that reduce excessive scarring and fibrous tissue accumulation as well as other related advantages.
  • drug-coated or drug- impregnated implants and medical devices which reduce fibrosis in the tissue surrounding the implanted drug delivery pump or sensor, or inhibit scar development on the device/implant surface (particularly the drug delivery catheter lumen and the sensor surface), thus enhancing the efficacy of the procedure.
  • fibrous tissue can reduce or obstruct the flow of therapeutic agents from the catheter to the target tissue, or prevent the implanted sensor from detecting accurate readings.
  • fibrosis is inhibited by local or systemic release of specific pharmacological agents that become localized to the tissue adjacent to the implanted device.
  • the repair of tissues following a mechanical or surgical intervention involves two distinct processes: (1) regeneration (the replacement of injured cells by cells of the same type and (2) fibrosis (the replacement of injured cells by connective tissue).
  • regeneration the replacement of injured cells by cells of the same type
  • fibrosis the replacement of injured cells by connective tissue.
  • connective tissue cells such as fibroblasts or smooth muscle cells
  • ECM extracellular matrix
  • angiogenesis formation of new blood vessels
  • remodeling maturation and organization of the fibrous tissue.
  • inhibitors (reduces) fibrosis may be understood to refer to agents or compositions which decrease or limit the formation of fibrous tissue (i.e., by reducing or inhibiting one or more of the processes of inflammation, connective tissue cell migration or proliferation, ECM production, angiogenesis, and/or remodeling).
  • numerous therapeutic agents described in this invention will have the additional benefit of also reducing tissue regeneration where appropriate.
  • an implant or device e.g., a sensor or pump
  • an implant or device is adapted to release an agent that inhibits fibrosis through one or more of the mechanisms cited herein.
  • an implant or device contains an agent that while remaining associated with the implant or device, inhibits fibrosis between the implant or device and the tissue where the implant or device is placed by direct contact between the agent and the tissue surrounding the implant or device.
  • implanted pumps and sensors are provided comprising an implant or device, wherein the implant or device releases an agent which inhibits fibrosis in vivo.
  • Release of an agent refers to any statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the implant/device and/or remains active on the surface of (or within) the device/implant.
  • methods are provided for manufacturing a medical device or implant, comprising the step of coating (e.g., spraying, dipping, wrapping, or administering drug through) a medical device or implant.
  • the implant or medical device can be constructed so that the device itself is comprised of materials which inhibit fibrosis in or around the implant.
  • implantable pumps and sensors may be utilized within the context of the present invention, depending on the site and nature of treatment desired.
  • the implanted pump or sensor is further coated with a composition or compound, which delays the onset of activity of the fibrosis-inhibiting agent for a period of time after implantation.
  • a composition or compound which delays the onset of activity of the fibrosis-inhibiting agent for a period of time after implantation.
  • agents include heparin, PLGA/MePEG, PLA, and polyethylene glycol.
  • the fibrosis-inhibiting implant or device is activated before, during, or after deployment (e.g., an inactive agent on the device is first activated to one that reduces or inhibits an in vivo fibrotic reaction).
  • the tissue surrounding the implanted pump (particularly the drug delivery catheter) and/or sensor is treated with a composition or compound that contains an inhibitor of fibrosis.
  • a composition or compound that contains an inhibitor of fibrosis e.g., topicals, injectables, liquids, gels, sprays, microspheres, pastes, wafers
  • compounds containing an inhibitor of fibrosis are described that can be applied to the surface of, or infiltrated into, the tissue adjacent to the pump or sensor, such that the pharmaceutical agent is delivered in therapeutic levels over a period sufficient to prevent the drug delivery catheter and/or sensor from being obstructed or encapsulated by fibrous tissue.
  • fibrosis-inhibitor This can be done in lieu of coating the device or implant with a fibrosis-inhibitor, or done in addition to coating the device or implant with a fibrosis-inhibitor.
  • the local administration of the fibrosis-inhibiting agent can occur prior to, during, or after implantation of the pump or sensor itself.
  • an implanted pump or sensor is coated on one aspect, portion or surface with a composition which inhibits fibrosis, as well as being coated with a composition or compound which promotes scarring on another aspect, portion or surface of the device (i.e., to affix the body of the device into a particular anatomical space).
  • agents that promote fibrosis and scarring include silk, silica, crystalline silicates, bleomycin, quartz dust, neomycin, talc, metallic beryllium and oxides thereof, retinoic acid compounds, copper, leptin, growth factors, a component of extracellular matrix; fibronectin, collagen, fibrin, or fibrinogen, polylysine, poly(ethylene-co-vinylacetate), chitosan, N-carboxybutylchitosan, and RGD proteins; vinyl chloride or a polymer of vinyl chloride; an adhesive selected from the group consisting of cyanoacrylates and crosslinked poly(ethylene glycol) - methylated collagen; an inflammatory cytokine (e.g., TGF ⁇ , PDGF, VEGF, bFGF, TNF ⁇ , NGF, GM-CSF, IGF-1 , IL-1 , IL-1- ⁇ , IL-8, IL- 6, and growth hormone); connective tissue growth factor (CT
  • Also provided by the present invention are methods for treating patients undergoing surgical, endoscopic or minimally invasive therapies where an implanted pump or sensor is placed as part of the procedure.
  • inhibits fibrosis refers to a statistically significant decrease in the amount of scar tissue in or around the device or an improvement in the interface between the implant (catheter and/or sensor) and the tissue, which may or may not lead to a permanent prohibition of any complications or failures of the device/implant.
  • the pharmaceutical agents and compositions are utilized to create novel drug-coated implants and medical devices that reduce the foreign body response to implantation and limit the growth of reactive tissue on the surface of, into, or around the device, such that performance is enhanced.
  • Implantable pumps and sensors coated with selected pharmaceutical agents designed to prevent scar tissue overgrowth and improve electrical conduction can offer significant clinical advantages over uncoated devices.
  • the present invention is directed to implantable pumps and sensors that comprise a medical implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti- scarring agent.
  • the agent is present so as to inhibit scarring that may otherwise occur when the implant is placed within an animal.
  • the present invention is directed to methods wherein both an implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti-scarring agent, are placed into an animal, and the agent inhibits scarring that may otherwise occur.
  • the present invention provides a device, comprising an implantable pump and/or sensor and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring.
  • the agent may be present in a composition along with a polymer.
  • the polymer is biodegradable.
  • the polymer is nonbiodegradable.
  • the present invention also provides methods.
  • the present invention provides methods whereby a specified device is implanted into an animal, and a specified agent associated with the device inhibits scarring that may otherwise occur.
  • the devices identified herein may be a "specified device”
  • each of the anti-scarring agents identified herein may be an "anti- scarring agent” where the present invention provides, in independent embodiments, for each possible combination of the device and the agent.
  • the agent may be associated with the device prior to the device being placed within the animal.
  • the agent or composition comprising the agent
  • the agent may be coated onto an implant, and the resulting device then placed within the animal.
  • the agent may be independently placed within the animal in the vicinity of where the device is to be, or is being, placed within the animal.
  • the agent may be sprayed or otherwise placed onto, adjacent to, and/or within the tissue that will be contacting the medical implant or may otherwise undergo scarring.
  • the present invention provides placing an implantable pump and/or sensor and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
  • the agent may be present in a composition along with a polymer.
  • the polymer is biodegradable.
  • the polymer is non-biodegradable.
  • the anti-fibrotic agent may be one or more of the following: 1) an anti-fibrotic agent that inhibits cell regeneration, 2) an anti-fibrotic agent that inhibits angiogenesis, 3) an anti-fibrotic agent that inhibits fibroblast migration, 4) an anti-fibrotic agent that inhibits fibroblast proliferation, 5) an anti-fibrotic agent that inhibits deposition of extracellular matrix, 6) an anti-fibrotic agent inhibits tissue remodeling, 7) an adensosine A2A receptor antagonist, 8) an AKT inhibitor, 9) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is
  • Pharmaprojects No. 5754 (Merck KgaA), 10) an alpha 4 integrin antagonist, 11) an alpha 7 nicotinic receptor agonist, 12) an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Se
  • a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof, 39) an endotoxin antagonist, 40) an epothilone and tubulin binder, 41) an estrogen receptor antagonist, 42) an FGF inhibitor, 43) a famexyl transferase inhibitor, 44) a farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a
  • FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof, 45) an FLT-3 kinase inhibitor, 46a) an FGF receptor kinase inhibitor, 47) a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No.
  • a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17- dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No.
  • an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No.
  • an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
  • autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No.
  • 334476- 64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof, 75) an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No.
  • INDRA OXiGENE
  • IPL- 576092 CAS No. 137571-30-3
  • Inflazyme NFKB decoy
  • NFKB decoy oligo NFKB decoy oligo
  • S5 F005 from Fulcrum Pharmaceuticals
  • P61 Physicaltopharm
  • R-flurbiprofen CAS No.
  • a palmitoyl-protein thioesterase inhibitor 80) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No.
  • a peroxisome proliferators-activated receptor agonist selected from the group consisting of (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No.
  • 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No.
  • MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L- 165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof, 82) a phosphatase inhibitor, 83) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No.
  • PDE phosphodiesterase
  • a phosphodiesterase III inhibitor enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof
  • a PKC inhibitor 85) a platelet activating factor antagonist, 86) a platelet-derived growth factor receptor kinase inhibitor, 87) a prolyi hydroxylase inhibitor, 88) a polymorphonuclear neutrophil inhibitor, 89) a protein kinase B inhibitor, 90) a protein kinase C stimulant, 91) a purine nucleoside analogue, 92) a purinoreceptor P2X antagonist, 93) a Raf kinase inhibitor, 94) a reversible inhibitor of ErbB1 and ErbB2, 95) a ribonucleoside triphosphate reductase inhibitor, 96
  • TNF ⁇ antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No.
  • simvastatin CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No.
  • brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetid ⁇ ne (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No.
  • H3 receptor antagonists e.g., thioperamide and thioperamide maleate salt
  • anti-histamines e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones
  • 131 an alpha adrenergic receptor antagonist
  • 132) an anti-psychotic compound
  • 133) a CaM kinase Il inhibitor
  • 134) a G protein agonist 135) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No.
  • a DNA topoisomerase inhibitor selected from the group consisting of ⁇ -lapachone (CAS No. 4707-32- 8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof, 138) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No.
  • a D2 dopamine receptor antagonist 140) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, 141) a dopamine antagonist, an anesthetic compound, 142) a clotting factor, 143) a lysyl hydrolase inhibitor, 144) a muscarinic receptor inhibitor, 145) a superoxide anion generator, 146) a steroid, 147) an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No.
  • temozolomide temozolomide
  • procarbazine HCl and an analogue or derivative thereof
  • 157) a DNA methylation inhibitor 158) a NSAID agent, 159) a peptidylglycine alpha- hydroxylating monooxygenase inhibitor, 160) an MEK1/MEK 2 inhibitor, 161) a NO synthase inhibitor, 162) a retinoic acid receptor antagonist selected from isotretinoin (CAS No.
  • an ACE inhibitor 164) a glycosylation inhibitor, 165) an intracellular calcium influx inhibitor, 166) an anti-emetic agent, 167) an acetylcholinesterase inhibitor, 168) an ALK-5 receptor antagonist, 169) a RAR/RXT antagonist, 170) an elF-2a inhibitor, 171) an S-adenosyl-L-homocysteine hydrolase inhibitor, 172) an estrogen agonist, 173) a serotonin receptor inhibitor, 174) an antithrombotic agent, 175) a tryptase inhibitor, 176) a pesticide, 177) a bone mineralization promoter, 178) a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, 179) an anti-inflammatory compound, 180) a DNA methylation promoter, 181) an anti-spasmodic agent, 182) a protein synthesis inhibitor, 183) an ⁇ -glucosidase inhibitor,
  • Figure 1A schematically depicts the transcriptional regulation of matrix metalloproteinases.
  • Figure 1B is a blot which demonstrates that IL-1 stimulates AP-1 transcriptional activity.
  • Figure 1C is a graph which shows that IL-1 induced binding activity decreased in lysates from chondrocytes which were pretreated with paclitaxel.
  • Figure 1 D is a blot which shows that IL-1 induction increases collagenase and stromelysin in RNA levels in chondrocytes, and that this induction can be inhibited by pretreatment with paclitaxel.
  • Figures 2A-H are blots that show the effect of various anti- microtubule agents in inhibiting collagenase expression.
  • Figure 3 is a graph showing the results of a screening assay for assessing the effect of paclitaxel on smooth muscle cell migration.
  • Figure 4 is a bar graph showing the area of granulation tissue in carotid arteries exposed to silk coated perivascular polyurethane (PU) films relative to arteries exposed to uncoated PU films.
  • PU perivascular polyurethane
  • Figure 5 is a bar graph showing the area of granulation tissue in carotid arteries exposed to silk suture coated perivascular PU films relative to arteries exposed to uncoated PU films.
  • Figure 6 is a bar graph showing the area of granulation tissue in carotid arteries exposed to natural and purified silk powder and wrapped with perivascular PU film relative to a control group in which arteries are wrapped with perivascular PU film only.
  • Figure 7 is a bar graph showing the area of granulation tissue (at 1 month and 3 months) in carotid arteries sprinkled with talcum powder and wrapped with perivascular PU film relative to a control group in which arteries are wrapped with perivascular PU film only.
  • Medical device “implant”, “device”, “medical device,” “medical implant”, “implant/device”, and the like are used synonymously to refer to any object that is designed to be placed partially or wholly within a patient's body for one or more therapeutic or prophylactic purposes such as for restoring physiological function, alleviating symptoms associated with disease, delivering therapeutic agents, detecting changes (or levels) in the internal environment, and/or repairing or replacing or augmenting etc. damaged or diseased organs and tissues.
  • medical devices are normally composed of biologically compatible synthetic materials (e.g., medical-grade stainless steel, titanium and other metals; exogenous polymers, such as polyurethane, silicon, PLA, PLGA), other materials may also be used in the construction of the medical device or implant.
  • Specific medical devices and implants that are particularly useful for the practice of this invention include devices and implants designed to deliver therapeutic levels of a drug to a target tissue (drug delivery pumps) and/or sensors designed to detect changes in body function and/or levels of key physiological metabolites, chemistry, hormones or biological factors.
  • Implantable sensor refers to a medical device that is implanted in the body to detect blood or tissue levels of a particular chemical (e.g., glucose, electrolytes, drugs, hormones) and/or changes in body chemistry, metabolites, function, pressure, flow, physical structure, electrical activity or other variable parameter.
  • Implantable sensors may have one or more electrodes that extend into the external environment to sense a variety of physical and/or physiological properties, including, but not limited to, optical, mechanical, baro, chemical and electrochemical properties. Sensors may be used to detect information, for example, about temperature, strain, pressure, magnetic, acceleration, ionizing radiation, acoustic wave or chemical changes (e.g., blood constituents, such as glucose).
  • the sensor may utilize an enzyme-based electrochemical sensor, a glucose-responsive hydrogel combined with a pressure sensor, microwires with electrodes, radiofrequency microelectronics and a glucose affinity polymer combined with physical and biochemical sensor technology, and near or mid infrared light emission combined with optical spectroscopy detectors to name a few.
  • implantable sensors include, blood/tissue glucose monitors, electrolyte sensors, blood constituent sensors, temperature sensors, pH sensors, optical sensors, amperometric sensors, pressure sensors, biosensors, sensing transponders, strain sensors, activity sensors and magnetoresistive sensors.
  • Drug-delivery pump refers to a medical device that includes a pump which is configured to deliver a biologically active agent (e.g., a drug) at a regulated dose. These devices are implanted within the body and may include an external transmitter for programming the controlled release of drug, or alternatively, may include an implantable sensor that provides the trigger for the drug delivery pump to release drug as physiologically required. Drug-delivery pumps may be used to deliver virtually any agent, but specific examples include insulin for the treatment of diabetes, medication for the relief of pain, chemotherapy for the treatment of cancer, anti-spastic agents for the treatment of movement and muscular disorders, or antibiotics for the treatment of infections.
  • a biologically active agent e.g., a drug
  • constant flow drug delivery pumps e.g., programmable drug delivery pumps, intrathecal pumps, implantable insulin delivery pumps, implantable osmotic pumps, ocular drug delivery pumps and implants
  • metering systems e.g., peristaltic (roller) pumps
  • electronically driven pumps elastomeric pumps
  • spring-contraction pumps e.g., gas-driven
  • Fibrosis refers to the formation of fibrous (scar) tissue in response to injury or medical intervention.
  • Therapeutic agents which inhibit fibrosis or scarring can do so through one or more mechanisms including: inhibiting the inflammatory response, inhibiting migration or proliferation of connective tissue cells (such as fibroblasts, smooth muscle cells, and vascular smooth muscle cells), inhibiting angiogenesis, reducing ECM production (or promoting ECM breakdown), and/or inhibiting tissue remodeling.
  • connective tissue cells such as fibroblasts, smooth muscle cells, and vascular smooth muscle cells
  • angiogenesis such as fibroblasts, smooth muscle cells, and vascular smooth muscle cells
  • reducing ECM production or promoting ECM breakdown
  • tissue remodeling reducing tissue remodeling
  • Inhibit fibrosis "reduce fibrosis”, “fibrosis-inhibitor”, “inhibits scar”, “reduces scar”, “anti-fibrosis”, “anti-fibrotic,” “anti-scarring” and the like are used synonymously to refer to the action of agents or compositions which result in a statistically significant decrease in the formation of fibrous tissue that may be expected to occur in the absence of the agent or composition.
  • “Inhibitor” refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process.
  • the process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
  • Antagonist refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process. While the process may be a general one, typically this refers to a drug mechanism where the drug competes with a molecule for an active molecular site or prevents a molecule from interacting with the molecular site. In these situations, the effect is that the molecular process is inhibited.
  • Antist refers to an agent which stimulates a biological process or rate or degree of occurrence of a biological process.
  • the process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
  • Anti-microtubule agents may be understood to include any protein, peptide, chemical, or other molecule which impairs the function of microtubules, for example, through the prevention or stabilization of polymerization.
  • Compounds that stabilize polymerization of microtubules are referred to herein as "microtubule stabilizing agents.”
  • a wide variety of methods may be utilized to determine the anti-microtubule activity of a particular compound, including for example, assays described by Smith et al. (Cancer Lett 79(2):213-219, 1994) and Mooberry et al., ⁇ Cancer Lett. 96(2):261- 266, 1995).
  • “Host”, “person”, “subject”, “patient” and the like are used synonymously to refer to the living being (human or animal) into which a device of the present invention is implanted. "Implanted” refers to having completely or partially placed a device within a host. A device is partially implanted when some of the device reaches, or extends to the outside of, a host.
  • Release of an agent refers to a statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the implant/device.
  • Biodegradable refers to materials for which the degradation process is at least partially mediated by, and/or performed in, a biological system.
  • Degradation refers to a chain scission process by which a polymer chain is cleaved into oligomers and monomers. Chain scission may occur through various mechanisms, including, for example, by chemical reaction (e.g., hydrolysis) or by a thermal or photolytic process.
  • Polymer degradation may be characterized, for example, using gel permeation chromatography (GPC), which monitors the polymer molecular mass changes during erosion and drug release.
  • GPC gel permeation chromatography
  • Biodegradable also refers to materials may be degraded by an erosion process mediated by, and/or performed in, a biological system.
  • Erosion refers to a process in which material is lost from the bulk.
  • the material may be a monomer, an oligomer, a part of a polymer backbone, or a part of the polymer bulk.
  • Erosion includes (i) surface erosion, in which erosion affects only the surface and not the inner parts of a matrix; and (ii) bulk erosion, in which the entire system is rapidly hydrated and polymer chains are cleaved throughout the matrix.
  • erosion generally occurs by one of three basic mechanisms (see, e.g., Heller, J., CRC Critical Review in Therapeutic Drug Carrier Systems (1984), 1(1), 39- 90); Siepmann, J.
  • analogue refers to a chemical compound that is structurally similar to a parent compound, but differs slightly in composition (e.g., one atom or functional group is different, added, or removed).
  • the analogue may or may not have different chemical or physical properties than the original compound and may or may not have improved biological and/or chemical activity.
  • the analogue may be more hydrophilic or it may have altered reactivity as compared to the parent compound.
  • the analogue may mimic the chemical and/or biologically activity of the parent compound (i.e., it may have similar or identical activity), or, in some cases, may have increased or decreased activity.
  • the analogue may be a naturally or non- naturally occurring (e.g., recombinant) variant of the original compound.
  • An example of an analogue is a mutein (i.e., a protein analogue in which at least one amino acid is deleted, added, or substituted with another amino acid).
  • Other types of analogues include isomers (enantiomers, diasteromers, and the like) and other types of chiral variants of a compound, as well as structural isomers.
  • the analogue may be a branched or cyclic variant of a linear compound.
  • a linear compound may have an analogue that is branched or otherwise substituted to impart certain desirable properties (e.g., improve hydrophilicity or bioavailability).
  • derivative refers to a chemically or biologically modified version of a chemical compound that is structurally similar to a parent compound and (actually or theoretically) derivable from that parent compound.
  • a “derivative” differs from an “analogue” in that a parent compound may be the starting material to generate a "derivative,” whereas the parent compound may not necessarily be used as the starting material to generate an “analogue.”
  • a derivative may or may not have different chemical or physical properties of the parent compound. For example, the derivative may be more hydrophilic or it may have altered reactivity as compared to the parent compound.
  • Derivatization may involve substitution of one or more moieties within the molecule (e.g., a change in functional group).
  • a hydrogen may be substituted with a halogen, such as fluorine or chlorine, or a hydroxyl group (-OH) may be replaced with a carboxylic acid moiety (-COOH).
  • derivative also includes conjugates, and prodrugs of a parent compound (i.e., chemically modified derivatives which can be converted into the original compound under physiological conditions).
  • the prodrug may be an inactive form of an active agent. Under physiological conditions, the prodrug may be converted into the active form of the compound.
  • Prodrugs may be formed, for example, by replacing one or two hydrogen atoms on nitrogen atoms by an acyl group (acyl prodrugs) or a carbamate group (carbamate prodrugs). More detailed information relating to prodrugs is found, for example, in Fleisher et al., Advanced Drug Delivery Reviews 19 (1996) 115; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; or H. Bundgaard, Drugs of the Future 16 (1991) 443.
  • the term "derivative” is also used to describe all solvates, for example hydrates or adducts (e.g., adducts with alcohols), active metabolites, and salts of the parent compound.
  • acidic groups for example carboxylic acid groups
  • alkali metal salts or alkaline earth metal salts e.g., sodium salts, potassium salts, magnesium salts and calcium salts
  • physiologically tolerable quaternary ammonium ions and acid addition salts with ammonia and physiologically tolerable organic amines such as, for example, triethylamine, ethanolamine or tris-(2-hydroxyethyl)amine.
  • Basic groups can form acid addition salts, for example with inorganic acids such as hydrochloric acid, sulfuric acid or phosphoric acid, or with organic carboxylic acids and sulfonic acids such as acetic acid, citric acid, benzoic acid, maleic acid, fumaric acid, tartaric acid, methanesulfonic acid or p-toluenesulfonic acid.
  • Compounds which simultaneously contain a basic group and an acidic group for example a carboxyl group in addition to basic nitrogen atoms, can be present as zwitterions.
  • Salts can be obtained by customary methods known to those skilled in the art, for example by combining a compound with an inorganic or organic acid or base in a solvent or diluent, or from other salts by cation exchange or anion exchange.
  • concentration ranges, percentage range, or ratio range recited herein are to be understood to include concentrations, percentages or ratios of any integer within that range and fractions thereof, such as one tenth and one hundredth of an integer, unless otherwise indicated.
  • any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness are to be understood to include any integer within the recited range, unless otherwise indicated.
  • the terms “a” and “an” as used above and elsewhere herein refer to “one or more” of the enumerated components.
  • “a” polymer refers to one polymer or a mixture comprising two or more polymers.
  • the term “about” means ⁇ 15%.
  • the present invention provides compositions, methods and devices relating to medical devices and implants (specifically implantable pumps and sensors), which greatly increase their ability to inhibit the formation of reactive scar tissue on, or around, the surface of the device or implant. Described in more detail below are methods for constructing medical devices or implants, compositions and methods for generating medical devices and implants which inhibit fibrosis, and methods for utilizing such medical devices and implants.
  • implantable sensors that include an anti-scarring agent are provided that can be used to detect physiological levels or changes in the body.
  • sensor devices where the occurrence of a fibrotic reaction will adversely affect the functioning of the device or the biological problem for which the device was implanted or used.
  • Proper clinical functioning of an implanted sensor is dependent upon intimate anatomical contact with the target tissues and/or body fluids. Scarring around the implanted device may degrade the electrical components and characteristics of the device-tissue interface, and the device may fail to function properly.
  • scar tissue between the sensing device and the adjacent (target) tissue can prevent the flow of physical, chemical and/or biological information (e.g., fluid levels, drug levels, metabolite levels, glucose levels, pressure etc.) from reaching the detection mechanism of the sensor.
  • biological information e.g., fluid levels, drug levels, metabolite levels, glucose levels, pressure etc.
  • the sensor is detecting conditions inside the capsule (i.e., levels detected in a microenvironment), and these conditions are not consistent with those outside the capsule (i.e., within the body as a whole - the microenvironment), it will record information that is not representative of systemic levels.
  • Sensors or transducers may be located deep within the body for monitoring a variety of physiological properties, such as temperature, pressure, strain, fluid flow, metabolite levels (e.g., electrolytes, glucose), drug levels, chemical properties, electrical properties, magnetic properties, and the like.
  • physiological properties such as temperature, pressure, strain, fluid flow, metabolite levels (e.g., electrolytes, glucose), drug levels, chemical properties, electrical properties, magnetic properties, and the like.
  • Representative examples of implantable sensors for use in the practice of the invention include, blood and tissue glucose monitors, electrolyte sensors, blood constituent sensors, temperature sensors, pH sensors, optical sensors, amperometric sensors, pressure sensors, biosensors, sensing transponders, strain sensors, activity sensors and magnetoresistive sensors.
  • the implantable sensor may be a microelectronic device that is implanted around the large bowels to control bowel function by detecting rectal contents and stimulating peristaltic contractions to empty the bowels when it is convenient. See, e.g., U.S. Patent No. 6,658,297.
  • the implantable sensor may be used to measure pH in the Gl tract.
  • a representative example of such a pH sensing device is the BRAVO pH Monitoring System from Medtronic, Inc. (Minneapolis, MN).
  • the implantable sensor may be part of a Gl catheter or probe that includes a sensor portion connected to an electrical or optical measurement device and a sensitive polymeric material that undergoes an irreversible change when exposed to cumulative action of an external medium. See, e.g., U.S. Patent No. 6,006,121.
  • the implantable sensor may be a component of a central venous catheter (CVC) (e.g., a jugular vein catheter) system.
  • the device may be composed of a catheter body having at least one oxygen sensor and a distal heat exchange region in which the catheter body is formed with coolant supply and return lumens to provide heat exchange within a body to prevent overheating due to severe brain trauma or ischemia due to stroke. See, e.g., U.S. Patent No. 6,652,565.
  • a CVC may include a thermal mass and a temperature sensor to measure blood temperature. See, e.g., U.S. Patent No. 6,383,144.
  • Glucose monitors are used to detect changes in blood glucose, specifically for the management and treatment of patients with diabetes mellitus. Diabetes is a metabolic disorder of glucose metabolism that afflicts tens of millions of people in the developed countries of the world. This disease is characterized by the inability of the body to properly utilize and metabolize carbohydrates, particularly glucose. Normally, the finely-tuned balance between glucose in the blood and glucose in the bodily tissue cells is maintained by insulin, a hormone produced by the pancreas.
  • pancreas becomes defective and insulin is produced in inadequate amounts to reduce blood glucose levels (Type I diabetes), or if the body becomes insensitive to the glucose-lowering effects of insulin despite adequate pancreatic insulin production (Type Il diabetes), the result is diabetes.
  • Accurate detection of blood glucose levels is essential to the management of diabetic patients because the dosage and timing of administration of insulin and/or other hypoglycemic agents are titrated depending upon changes in glucose levels in response to the medication. If the dosage is too high, blood glucose levels drop too low, resulting in confusion and potentially even loss of consciousness. If the dosage is too low, blood glucose levels rise too high, leading to excessive thirst, urination, and changes in metabolism known as ketoacidosis.
  • glucose levels can fluctuate wildly between the two extremes - a situation that is thought to contribute to some of the long-term complications of diabetes such as heart disease, kidney failure and blindness. Since in the extreme, all these conditions can be life threatening, careful and continuous monitoring of glucose levels is a critical aspect of diabetes management.
  • One way to detect changes in glucose levels and to continuously sense when levels of glucose become too high or too low in diabetes patients is to implant a glucose sensor. As the glucose sensor detects changes in the blood glucose levels, insulin can be administered by external injection or via an implantable insulin pump to maintain blood glucose levels within an acceptable physiologic range. Numerous types of blood and tissue glucose monitors are suitable for use in the practice of the invention.
  • the glucose monitor may be delivered to the vascular system transluminal ⁇ using a catheter on a stent platform.
  • the glucose monitor may be composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations. See, e.g., U.S. Patent Nos. 5,101 ,814 and 5,190,041.
  • the glucose monitor may be a small diameter flexible electrode implanted subcutaneously which may be composed of an analyte-responsive enzyme designed to be an electrochemical glucose sensor. See, e.g., U.S. Patent Nos. 6,121 ,009 and 6,514,718.
  • the implantable sensor may be a closed loop insulin delivery system whereby there is a sensing means that detects the patient's blood glucose level based on electrical signals and then stimulates either an insulin pump or the pancreas to supply insulin. See, e.g., U.S. Patent Nos. 6,558,345 and 6,093,167. Other glucose monitors are described in, for e.g., U.S. Patent Nos. 6,579,498; 6,565,509 and 5,165,407. Minimally invasive glucose monitors include the GLUCOWATCH G2 BIOGRAPHER from Cygnus Inc. (see cygn.com); see, e.g., U.S. Patent Nos.
  • the CONTINUOUS GLUCOSE MONITORING SYSTEM from Medtronic MiniMed, Inc. (Northridge, CA; see minimed.com); see, e.g., U.S. Patent Nos. 6,520,326; 6,424,847; 6,360,888; 5,605,152; 6,804,544; and U.S. Patent Application No. 20040167464A1.
  • the CGMS system is surgically implanted in the subcutaneous tissue of the abdomen and stores tissue glucose readings every 5 minutes. Coating the sensor with a fibrosis-inhibiting agent may prolong the activity of this device because it often must be removed after several days (approximately 3), in part because it loses its sensitivity as a result of the local tissue reaction to the device.
  • the CONTINUOUS GLUCOSE MONITORING DEVICE from TheraSense (Alameda, CA, see therasense.com) which utilizes a disposable, miniaturized electrochemical sensor that is inserted under the patient's skin using a spring-loaded insertion device.
  • the sensor measures glucose levels in the interstitial fluid every five minutes, with the ability to store results for future analysis. See, e.g., US20040186365A1 ; US20040106858A1 and US20030176183A1. Even though the device can store up to a month of data and has alarms for high and low glucose levels, it must be replaced every few days because it loses its accuracy as a result of the foreign body reaction to the implant.
  • Another electrochemical sensor that may benefit from the present invention is the multilayered implantable electrochemical sensor from lsense (Portland, OR).
  • This system consists of a semipermeable membrane, a catalytic membrane which generates an electrical current in the presence of glucose, and a specificity membrane to reduce interference from other substances.
  • the SMSI glucose sensor (Sensors for Medicine and Sciences, Inc., Montgomery County, Maryland; see s4ms.com) is designed to be implanted under the skin in a short outpatient procedure.
  • the sensor is designed to automatically measure interstitial glucose every few minutes, without any user intervention.
  • the sensor implant communicates wirelessly with a small external reader, allowing the user to monitor glucose levels continuously or on demand.
  • the reader is designed to be able to track the rate of change of glucose levels and warn the user of impending hypo- or hyperglycemia.
  • the operational life of the sensor implant is about 6-12 months, after which it may be replaced.
  • Animas Corporation (West Chester, PA; animascorp.com) is developing an implantable glucose sensor that measures the near-infrared absorption of blood based on spectroscopy or optical sensing placed around a vein.
  • the Animas glucose monitor may be tied to an insulin infusion pump to provide a closed-loop control of blood glucose levels. Scar tissue over the sensor distorts the ability of the device to correctly gather optical information and may thus benefit from use in combination with a fibrosis inhibiting agent.
  • DexCom, Inc. (San Diego, CA; see dexcom.com) is developing their Continuous Glucose Monitoring System which is an implantable sensor that wirelessly transmits continuous blood glucose readings to an external receiver.
  • the receiver displays the current glucose value every 30 seconds, as well as one-hour, three-hour and nine-hours trended values, and sounds an alert when a high or low glucose excursion is detected.
  • This device features an implantable sensor that is placed in the subcutaneous tissue and continuously monitors tissue (interstitial fluid) glucose levels for both type 1 and type 2 diabetics.
  • This device may also include a unique microarchitectural arrangement in the sensor region that allows accurate data to be obtained over long periods of time.
  • Glucose monitoring devices and associated systems that are developed by DexCom, Inc. are described in, for example, U.S. Patent Nos. 6,741 ,877; 6,702,857 and 6,558,321.
  • glucose monitoring systems that utilize a glucose-responsive polymer as part of their detection mechanism.
  • M-Biotech (Salt Lake City, UT) is developing a continuous monitoring system that consists of subcutaneous implantation of a glucose-responsive hydrogel combined with a pressure transducer. See, e.g., U.S. Patent Nos.; and.
  • the hydrogel responds to changes in glucose concentration by either shrinking or swelling and the expansion or contraction is detected by the pressure transducer.
  • the transducer converts the information into an electrical signal and sends a wireless signal to a display device.
  • Cybersensors (Berkshire, UK) produces a capsule-like sensor implanted under the skin and an external receiver/transmitter that captures the data and powers the capsule via RF signals (see, e.g., GB 2335496 and U.S. Patent No. 6,579,498) Issued by the UK Patent and Trademark Office).
  • the sensor capsule is composed of a glucose affinity polymer and contains a physical sensor and an RF microchip; the entire capsule is further enclosed in a semipermeable membrane.
  • the glucose affinity polymer exhibits rheological changes when exposed to glucose (in the range of 3-15 nM) by becoming thinner and less viscous as glucose concentrations increase. This reversible reaction can be detected by the physical sensor and converted into a signal.
  • Advanced Biosensors that consists of small (150 ⁇ m wide by 2 mm long), biocompatible, silicon-based needles that are implanted under the skin.
  • the device senses glucose levels in the dermis and transmits data wirelessly.
  • a foreign body response and/or encapsulation of the implant affect the ability of the device to detect glucose levels accurately for longer than 7 days.
  • Combining this device with an inhibitor of fibrosis may allow it to accurately detect glucose levels for longer periods of time and extend the effective lifespan of the device.
  • the device must be accurately positioned adjacent to the tissue.
  • the detector of the sensing mechanism must be exposed to glucose levels that are identical to (or representative of) those found in the bloodstream. If excessive scar tissue growth or extracellular matrix deposition occurs around the device, this can impair the movement of glucose from the tissue to the detector and render it ineffective. Similarly if a "foreign body" response occurs and causes the implanted glucose sensor to become encapsulated by fibrous tissue, the sensor will be detecting glucose levels in the capsule.
  • glucose levels inside the capsule are not consistent with those outside the capsule (i.e., within the body as a whole), it will record information that is not representative of systemic levels. This can cause the physician or the patient to administer the wrong dosage of hypoglycemic drugs (such as insulin) with potentially serious consequences.
  • Blood, tissue or interstitial fluid glucose sensor devices that release a therapeutic agent able to reduce scarring and/or encapsulation of the implant can increase the efficiency and accuracy of glucose detection, minimize insulin dosing errors, assist in the maintenance of correct blood glucose levels, increase the duration that these devices function clinically, and/or reduce the frequency of implant replacement.
  • the device includes blood, tissue and interstitial fluid glucose monitoring devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent.
  • the fibrosis-inhibiting agent can also be incorporated into, and released from, the components of the implanted sensor.
  • This embodiment is particularly useful for implants employing glucose-responsive polymers and hydrogels (that can be drug-loaded with an active agent) as well as those utilizing a semi-permeable membrane around the sensor (which can also be loaded with a fibrosis- inhibiting agent).
  • a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the glucose sensor is, or will be, implanted.
  • the implantable sensor may be a pressure monitor.
  • Pressure monitors may be used to detect increasing pressure or stress within the body.
  • Implantable pressure transducers and sensors are used for temporary or chronic use in a body organ, tissue or vessel for recording absolute pressure. Many different designs and operating systems have been proposed and placed into temporary or chronic use for patients with a variety of medical conditions. Indwelling pressure sensors for temporary use of a few days or weeks are available, however, chronically or permanently implantable pressure sensors have also been used.
  • Pressure sensors may detect many types of bodily pressures, such as, but not limited to blood pressure and fluid flow, pressure within aneurysm sacs, intracranial pressure, and mechanical pressure associated with bone fractures.
  • the implantable sensor may detect body fluid absolute pressure at a selected site and ambient operating temperature by using a lead, sensor module, sensor circuit (including electrical conductors) and means for providing voltage. See, e.g., U.S. Patent No. 5,535,752.
  • the implantable sensor may be an intracranial pressure monitor that provides an analogue data signal which is converted electronically to a digital pulse. See, e.g., U.S. Patent No. 6,533,733.
  • the implantable sensor may be a barometric pressure sensor enclosed in an air chamber which is used for deriving reference pressure data for use in combination with an implantable medical device, such as a pacemaker. See, e.g., U.S.
  • the implantable sensor may be adapted to be inserted into a body passageway to monitor a parameter related to fluid flow through an endoluminal implant (e.g., stent). See, e.g., U.S. Patent No. 5,967,986.
  • the implantable sensor may be a passive sensor with an inductor-capacitor circuit having a resonant frequency which is adapted for the skull of a patient to sense intracranial pressure. See, e.g., U.S. Patent No. 6,113,553.
  • the implantable sensor may be a self- powered strain sensing system that generates a strain signal in response to stresses that may be produced at a bone fixation device. See, e.g., U.S.
  • the implantable sensor may be a component of a perfusion catheter.
  • the catheter may include a wire electrode and a lumen for perfusing saline around the wire, which is designed for measuring a potential difference across the Gl wall and for simultaneous measurement of pressure. See, e.g., U.S. Patent No. 5,551 ,425.
  • the implantable sensor may be part of a CNS device; for example, an intracranial pressure sensor which is mounted within the skull of a body at the situs where the pressure is to be monitored and a means of transmitting the pressure externally from the skull. See, e.g., U.S. Patent No. 4,003,141.
  • the implantable sensor may be a component of a left ventricular assist device.
  • the VAD may be a blood pump adapted to be joined in flow communication between the left ventricle and the aorta using an inlet flow pressure sensor and a controller that may adjust speed of pump based on sensor feedback.
  • a controller that may adjust speed of pump based on sensor feedback.
  • Numerous commercially available and experimental pressure and stress sensor devices are suitable for the practice of the invention. By way of illustration, a selection of these devices and implants are described in the following paragraphs
  • a device from CardioMEMS (Atlanta, GA; @cardiomems.com, a partnership between the Georgia Institute of Technology and the Cleveland Clinic) which can be inserted into an aneurysm sac to monitor pressure within the sac and thereby alert a medical specialist to the filing of the sac with fluid, possibly to rupture-provoking levels.
  • Endovascular aneurysm repair (EVAR) is often performed using a stent graft which isolates the aneurysm from the circulation.
  • EVAR Endovascular aneurysm repair
  • the CardioMEMS device is implanted into the aneurysm sac after EVAR to monitor pressure in the isolated sac in order to detect which patients are at increasing risk of rupture.
  • the pressure sensor features an inductive-capacitive resonant circuit with a variable capacitor. Since capacitance varies with the pressure in the environment in which the capacitor is placed, it can detect changes in local pressure. Data is generated by using external excitation systems that induce an oscillating current in the sensor and detecting the frequency of oscillation (which is then used to calculate pressure). Unfortunately, even though the circuitry allows long-term functioning, a foreign body response and/or encapsulation of the implant affect the ability of the device to detect accurate pressure levels in the aneurysm (i.e., the device detects the pressure in the microenvironment of the capsule, not of the aneurysm sac as a whole).
  • Combining this device with an inhibitor of fibrosis may allow it to accurately detect pressure levels for longer periods of time after implantation and reduce the number of devices that fail.
  • MicroStrain Inc. (Williston, VT, @microstrain.com) has developed a family of wireless implantable sensors for measuring strain, position and motion within the body. These sensors can measure, for example, eye tremor, depth of corneal implant, orientation sensor for improved tooth crown prep, mayer ligament strains, spinal ligament strains, vertebral bone strains, elbow ligament strains, emg and ekg data, 3DM-G for measurement of orientation and motion, wrist ligament strains, hip replacement sensors for measuring micromotion, implant subsidence, knee ligament strain, ankle ligament strain, Achilles tendon strain, foot arch support strains, force within foot insoles. The company provides a knee prosthesis that can measure in vivo compressive forces and transmit the data in real time.
  • Patents describing this technology, and components used in the manufacture of devices for this technology include US 6,714,763; 6,625,517; 6,622,567; 6,588,282; 6,529,127; 6,499,368; 6,433,629; 5,887,351 ; 5,777,467; 5,497,147; and 4,993,428.
  • US Patent Applications describing this technology, and components used in the manufacture of devices for this technology include 20040113790;
  • CMOS-based sensor can be implanted during standard surgical procedures and is inductively linked to an external unit integrated into a spectacle frame.
  • the glasses are in turn linked via a cable to a portable data logger. Data is relayed upstream to the glasses using a modulated RF carrier operating at 13.56 MHz and a switchable load, while power comes downstream to the sensor.
  • a modulated RF carrier operating at 13.56 MHz and a switchable load
  • the pressure range to which the sensor responds can be adapted between 50kNm-2 and 3.5MNm-2.
  • the device consists of a fine, foldable coil for telemetric coupling and a very small miniaturized pressure sensor.
  • the sensor is manufactured on a micro- technological basis and serves for continuous, long-term reading and monitoring of intraocular pressure.
  • Chip and coil are integrated in modified soft intraocular lenses, which can be implanted in the patient's eye during today's common surgical procedures.
  • the device often fails after initially successful implantation because a foreign body response and/or encapsulation of the implant affect the ability of it to detect accurate pressure levels in the eye (Ae., the device detects the pressure in the microenvironment of the capsule surrounding the implant, not intraocular pressure as a whole).
  • Combining this device with an inhibitor of fibrosis e.g., by coating the implant and/or sensor with the agent, incorporating the agent into the polymers that make up the implant, and/or infiltrating it into the eye tissue surrounding the implant) may allow it to accurately detect pressure levels for longer periods of time after implantation and reduce the number of devices that fail.
  • the device for accurate detection of physical and/or physiological properties (such as pressure), the device must be accurately positioned within the tissue and receive information that is representative of conditions as a whole. If excessive scar tissue growth or extracellular matrix deposition occurs around the device, the sensor may receive erroneous information that compromises its efficacy or the scar tissue may block the flow of biological information to the sensor. For example, many devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant pressure levels (i.e., the device detects the pressure in the microenvironment of the capsule surrounding the implant, not the pressure of the larger environment).
  • the device includes implantable sensor devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent.
  • the fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted sensor.
  • a composition that includes an anti- scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
  • the implantable sensor may be a device configured to detect properties in the heart or in cardiac muscle tissue.
  • Cardiac sensors are used to detect parameters associated with the performance of the heart as monitored at any given time point along a prolonged time period.
  • monitoring of the heart is often conducted to detect changes associated with heart disease, such as chronic heart failure (CHF).
  • CHF chronic heart failure
  • monitoring patterns associated with heart function deterioration based on hemodynamic changes can be detected (parameters such as cardiac output, ejection fraction, pressure, ventricular wall motion, etc.).
  • This constant direct monitoring is central to disease management in patients that present with CHF.
  • a hemodynamic crisis can be detected and the appropriate medications and interventions selected.
  • Numerous types of cardiac sensors are suitable for use in the practice of the invention.
  • the implantable sensor may be an activity sensor incorporating a magnet and a magnetoresistive sensor that provides a variable activity signal as part of a cardiac device. See, e.g., U.S. Patent No. 6,430,440 and 6,411 ,849.
  • the implantable sensor may monitor blood pressure in a heart chamber by emitting wireless communication to a remote device. See, e.g., U.S. Patent No. 6,409,674.
  • the implantable sensor may be an accelerometer-based cardiac wall motion sensor which transduces accelerations of cardiac tissue to a cardiac stimulation device by using electrical signals. See, e.g., U.S. Patent No. 5,628,777.
  • the implantable sensor may be implanted in the heart's cavity with an additional sensor implanted in a blood vessel to detect pressure and flow within heart's cavity. See, e.g., U.S. Patent No. 6,277,078.
  • CARDIAC AIRBAG ICD SYSTEM is a rhythm monitoring device that offers rescue shock capability delivering 30 Joule shock therapies for up to 3 episodes of ventricular fibrillation. In addition to the rescue shock capability the system can also provide bradycardia pacing and VT monitoring.
  • the PROTOS family of pacemakers from Biotronik (see biotronikusa.com) also incorporates pacing sensor capability called Closed Loop Simulation. Blood flow and tissue perfusion monitors can be used to monitor noncard ⁇ ac tissue as well.
  • the device for accurate detection of physical and/or physiological properties (such as pressure, flow rates, etc.), the device must be accurately positioned within the heart muscle, chambers or great vessels and receive information that is representative of conditions as a whole. If excessive scar tissue growth or extracellular matrix deposition occurs around the sensing device, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of biological information to the detector mechanism of the sensor. For example, many cardiac monitoring devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (Ae., the device detects conditions in the microenvironment of the capsule surrounding the implant, not the pressure of the larger environment).
  • the device includes implantable sensor devices that are coated with an anti- scarring agent or a composition that includes an anti-scarring agent.
  • the fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted cardiac sensor.
  • a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
  • the implantable sensor may be a device configured to detect properties in the respiratory system.
  • Respiratory sensors may be used to detect changes in breathing patterns.
  • a respiratory sensor may be used to detect sleep apnea, which is an airway disorder.
  • sleep apnea There are two kinds of sleep apnea. In one condition, the body fails to automatically generate the neuromuscular stimulation necessary to initiate and control a respiratory cycle at the proper time. In the other condition, the muscles of the upper airway contract during the time of inspiration and thus the airway becomes obstructed.
  • the cardiovascular consequences of apnea include disorders of cardiac rhythm (bradycardia, auriculoventricular block, ventricular extrasystoles) and hemodynamic disorders (pulmonary and systemic hypertension).
  • implantable sensors may be used to monitor respiratory functioning to detect an apnea episode so the appropriate response (e.g., electrical stimulation to the nerves of the upper airway muscles) or other treatment can be provided.
  • the implantable sensor may be a respiration element implanted in the thoracic cavity which is capable of generating a respiration signal as part of a ventilation system for providing gas to a host.
  • the implantable sensor may be composed of a sensing element connected to a lead body which is inserted into bone (e.g., manubrium) that communicates with the intrathoracic cavity to detect respiratory changes. See, e.g., U.S. Patent No. 6,572,543.
  • the device for accurate detection of physical and/or physiological properties, the device must be accurately positioned adjacent to the tissue. If excessive scar tissue growth or extracellular matrix deposition occurs around the pulmonary function or airway sensing device, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of biological information to the detector mechanism of the sensor. For example, many pulmonary function sensing devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (i.e., the device detects conditions in the microenvironment of the capsule surrounding the implant, not the functioning of the respiratory system as whole).
  • Respiratory sensing devices that release a therapeutic agent able to reduce scarring can increase the efficiency of detection and increase the duration that these devices function clinically.
  • the device includes implantable sensor devices that are coated with an anti- scarring agent or a composition that includes an anti-scarring agent.
  • the fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted respiratory sensor.
  • a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
  • the implantable sensor may be a device configured to detect properties in the auditory system.
  • Auditory sensors are used as part of implantable hearing systems for rehabilitation of pure sensorineural hearing losses, or combined conduction and inner ear hearing impairments.
  • Hearing systems may include an implantable sensor which delivers an electrical signal which is processed by an implanted processor and delivered to an implantable electromechanical transducer which acts on the middle or inner ear.
  • the auditory sensor acts as the microphone of the hearing system and acts to convert the incident airborne sound into an electrical signal.
  • Numerous types of auditory sensors as part of a hearing system are suitable for use in the practice of the invention.
  • the implantable sensor may generate an electrical audio signal as part of a hearing system for rehabilitation of hearing loss. See, e.g., U.S. Patent No.
  • the implantable sensor may be a capacitive sensor which is mechanically or magnetically coupled to a vibrating auditory element, such as the malleus, which detects the time-varying capacitance values resulting from the vibrations. See, e.g., U.S. Patent No. 6,190,306.
  • the implantable sensor may be an electromagnetic sensor having a permanent magnet and a coil and a time- varying magnetic flux linkage based on the vibrations which are provided to an output stimulator for mechanical or electrical stimulation of the cochlea. See, e.g., U.S. Patent No. 5,993,376.
  • auditory sensor devices suitable for the practice of the invention include: the HIRES 9OK Bionic Ear Implant, HIRESOLUTION SOUND, CLARION CII Bionic Ear, and CLARION 1.2, from Advanced Bionics (Sylmar, California, a Boston Scientific Company, see advancedbionics.com); see also U.S. Patent Nos.
  • the device for accurate detection of sound, the device must be accurately positioned within the ear. If excessive scar tissue growth or extracellular matrix deposition occurs around the auditory sensor, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of sound waves to the detector mechanism of the sensor. Auditory sensing devices that release a therapeutic agent able to reduce scarring can increase the efficiency of sound detection and increase the duration that these devices function clinically.
  • the device includes implantable sensor devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent.
  • the fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted auditory sensor.
  • a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
  • implantable sensors may be used to detect electrolytes and metabolites in the blood.
  • the implantable sensor may be a device to monitor constituent levels of metabolites or electrolytes in the blood by emitting a source of radiation directed towards blood such that it interacts with a plurality of detectors that provide an output signal.
  • the implantable sensor may be a biosensing transponder which is composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader. See, e.g., U.S. Patent No. 5,833,603.
  • the implantable sensor may be a monolithic bioelectronic device for detecting at least one analyte within the body of an animal. See, e.g., U.S. Patent No. 6,673,596. Other sensors that measure chemical analytes are described in, e.g., U.S. Patent Nos. 6,625,479 and 6,201 ,980. If excessive scar tissue growth or extracellular matrix deposition occurs around the sensor, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of metabolites or electrolytes to the detector mechanism of the sensor.
  • the device includes implantable sensor devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent.
  • the fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted sensor.
  • a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
  • implantable sensor devices Although numerous examples of implantable sensor devices have been described above, all possess similar design features and cause similar unwanted foreign body tissue reactions following implantation. It may be obvious to one of skill in the art that commercial sensor devices not specifically cited above as well as next-generation and/or subsequently-developed commercial sensor products are to be anticipated and are suitable for use under the present invention.
  • the sensor device, particularly the sensing element must be positioned in a very precise manner to ensure that detection is carried out at the correct anatomical location in the body. All, or parts, of a sensor device can migrate following surgery, or excessive scar tissue growth can occur around the implant, which can lead to a reduction in the performance of these devices.
  • Implantable sensor devices that release a therapeutic agent for reducing scarring (or fibrosis) at the sensor-tissue interface can be used to increase the efficacy and/or the duration of activity of the implant.
  • the present invention provides implantable sensor devices that include an anti-scarring agent or a composition that includes an anti-scarring agent.
  • compositions can further include one or more fibrosis-inhibiting agents such that the overgrowth of granulation, fibrous, or neointimal tissue is inhibited or reduced.
  • Methods for incorporating fibrosis-inhibiting compositions onto or into these sensor devices include: (a) directly affixing to the sensing device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier), (b) directly incorporating into the sensing device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier (c) by coating the sensing device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving a fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the sensing device, (e) by inserting the sensing device into a sleeve or mesh which is comprised of, or coated with, a fibrosis-inhibiting composition, (f) constructing the sensing device itself (or a
  • the coating process can be performed in such a manner as to: (a) coat a portion of the sensing device (such as the detector); or (b) coat the entire sensing device with the fibrosis-inhibiting composition.
  • the fibrosis- inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final product.
  • a medical device may be prepared which has a coating, where the coating is, e.g., uniform, non-uniform, continuous, discontinuous, or patterned.
  • an implantable sensor device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug (Ae., one or more fibrosis-inhibiting agents).
  • the reservoirs may be formed from divets in the device surface or micropores or channels in the device body.
  • the reservoirs are formed from voids in the structure of the device.
  • the reservoirs may house a single type of drug (e.g., fibrosis-inhibiting agent) or more than one type of drug (e.g., a fibrosis-inhibiting agent and an anti-infective agent).
  • the drug(s) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs.
  • a carrier e.g., a polymeric or non-polymeric material
  • the filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier.
  • the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount (dose) of drug, and each layer may have a different composition to further tailor the amount and type of drug that is released from the substrate.
  • the multi-layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void.
  • the coating of the medical device may directly contact the implantable sensor device, or it may indirectly contact the device when there is something, e.g., a polymer layer, that is interposed between the sensor device and the coating that contains the fibrosis-inhibiting agent.
  • the fibrosis-inhibiting agent can be applied directly or indirectly to the tissue adjacent to the sensor device (preferably near the sensor-tissue interface).
  • the fibrosis-inhibiting agent with or without a polymeric, non- polymeric, or secondary carrier: (a) to the sensor and/or detector surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) prior to, immediately prior to, or during, implantation of the sensor; (c) to the surface of the sensor and/or the tissue surrounding the implanted sensor and/or detector (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the sensor; (d) by topical application of the anti-fibrosis agent into the anatomical space where the implantable sensor will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemul
  • polymeric carriers themselves can help prevent the formation of fibrous tissue on the sensor and/or fibrous encapsulation of the implanted sensor. These carriers (described below) are particularly useful for the practice of this embodiment, either alone, or in combination with a fibrosis-inhibiting composition.
  • the following polymeric carriers can be infiltrated (as described in the previous paragraph) into the vicinity of the sensor-tissue interface and include: (a) sprayable collagen- containing formulations such as COSTASIS and crosslinked derivatized poly(ethylene glycol) -collagen compositions (described, e.g., in U.S. Patent Nos.
  • CT3 both from Angiotech Pharmaceuticals, Inc., Canada
  • sprayable PEG-containing formulations such as COSEAL (Angiotech Pharmaceuticals, Inc.), FOCALSEAL (Genzyme Corporation, Cambridge, MA), SPRAYGEL or DURASEAL (both from Confluent Surgical, Inc., Boston, MA), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the detector/sensor surface);
  • fibrinogen- containing formulations such as FLOSEAL or TISSEAL (both from Baxter Healthcare Corporation, Fremont, CA), either alone, or loaded with a fibrosis- inhibiting agent, applied to the implantation site (or the detector/sensor surface);
  • hyaluronic acid-containing formulations such as RESTYLANE or PERLANE (both from Q-Med AB, Sweden), HYLAFORM (Inamed Corporation, Santa Barbara, CA), SYNVISC (Biomatrix, Inc., Ridgefield, NJ), SEPRAFILM or SEPRACOAT (both from Genzyme Corporation), loaded with a fibrosis- inhibiting agent applied to the implantation site (or the detector/sensor surface);
  • polymeric gels for surgical implantation such as REPEL (Life Medical Sciences, Inc., Princeton, NJ) or FLOWGEL (Baxter Healthcare Corporation) alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the detector/sensor surface);
  • orthopedic "cements” used to hold prostheses and tissues in place loaded with a fibrosis-inhibiting agent applied to the implantation site (or the detector/sensor surface) such as OSTEOBOND (Zimmer, Inc., Warsaw, IN), low viscosity cement (LVC) from Wright Medical Technology, Inc.
  • SIMPLEX P (Stryker Corporation, Kalamazoo, Ml), PALACOS (Smith & Nephew Corporation, United Kingdom), and ENDURANCE (Johnson & Johnson, Inc., New Brunswick, NJ); (g) surgical adhesives containing cyanoacrylates such as DERMABOND (Johnson & Johnson, Inc., New Brunswick, NJ), INDERMIL (U.S. Surgical Company, Norwalk, CT), GLUSTITCH (Blacklock Medical Products Inc., Canada), TlSSUMEND (Veterinary Products Laboratories, Phoenix, AZ), VETBOND (3M Company, St. Paul, MN), HISTOACRYL BLUE (Davis & Geek, St.
  • DERMABOND Johnson & Johnson, Inc., New Brunswick, NJ
  • INDERMIL U.S. Surgical Company, Norwalk, CT
  • GLUSTITCH Blacklock Medical Products Inc., Canada
  • TlSSUMEND (Veterinary Products Laboratories, Phoenix, AZ), VETBOND (3M Company
  • a preferred polymeric matrix which can be used to help prevent the formation of fibrous tissue on the sensor and/or fibrous encapsulation of the implanted sensor, either alone or in combination with a fibrosis inhibiting agent/composition is formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydrylj (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents.
  • reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra
  • Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents.
  • Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500.
  • collagen or a collagen derivative is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue around the implanted sensor.
  • collagen or a collagen derivative e.g., methylated collagen
  • any anti-scarring agent described below may be utilized alone, or in combination, in the practice of this embodiment.
  • the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined.
  • the fibrosis-inhibiting agents used alone or in combination, may be administered under the following dosing guidelines:
  • Therapeutic agents that may be used include but are not limited to: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglo
  • Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 . Minimum concentration of 10 "8 - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • Kinesin Antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 . Minimum concentration of 10 ⁇ - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • TNF alpha antagonists including etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg.
  • Minimum concentration of 10 " 8 - 10 '4 M of agent is to be maintained on the implant or barrier surface.
  • Minimum concentration of 10 ⁇ 8 - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • NF Kappa B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 . Minimum concentration of 10 " 8 - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • Minimum concentration of 10 "8 - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC 50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus.
  • selected assays described herein such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus.
  • the total dose typically should not exceed 200 mg (range of 0.1 ⁇ g to 200 mg) and preferably 1 ⁇ g to 100 mg; dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 ; and minimum concentration of 10 "8 - 10 "4 M of agent should be maintained on the implant or barrier surface.
  • agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC 50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate.
  • the total dose typically should not to exceed 500 mg (range of 1.0 ⁇ g to 500 mg) and preferably 1 ⁇ g to 200 mg; dose per unit area of 0.01 ⁇ g - 200 ⁇ g per mm 2 , preferably 0.1 ⁇ g/mm 2 - 40 ⁇ g/mm 2 ; and minimum concentration of 10 ⁇ - 10 "4 M of agent should be maintained on the implant or barrier surface.
  • agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC 50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
  • the total dose typically should not exceed 1000 mg (range of 0.1 ⁇ g to 1000 mg), preferably 1 ⁇ g to 500 mg; dose per unit area of 0.01 ⁇ g - 500 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 100 ⁇ g/mm 2 ; and minimum concentration of 10 '8 - 10 ⁇ 4 M of agent should to be maintained on the implant or barrier surface.
  • implantable pumps that include an anti-scarring agent that can be used to deliver drugs to a desired location.
  • Implantable drug delivery devices and pumps are a means to provide prolonged, site-specific release of a therapeutic agent for the management of a variety of medical conditions.
  • Drug delivery implants and pumps are generally utilized when a localized pharmaceutical impact is desired (i.e., the condition affects only a specific region) or when systemic delivery of the agent is inefficient or ineffective (Ae., leads to toxicity or severe side effects, results in inactivation of the drug prior to reaching the target tissue, produces poor symptom/disease control, and/or leads to addiction to the medication).
  • Implantable pumps can also deliver systemic drug levels in a constant, regulated manner for extended periods and help patients avoid the "peaks and valleys" of blood-level drug concentrations associated with intermittent systemic dosing.
  • Another advantage of implantable pumps is improved patient compliance. Many patients forget to take their medications regularly (particularly the young, elderly, chronically ill, mentally handicapped), but with an implantable pump, this problem is alleviated. For many patients this can lead to better symptom control (the dosage can often be titrated to the severity of the symptoms), superior disease management (particularly for insulin delivery in diabetics), and lower drug requirements (particularly for pain medications).
  • Innumerable drug delivery implants and pumps have been used in a variety of clinical applications, including programmable insulin pumps for the treatment of diabetes, intrathecal (in the spine) pumps to administer narcotics (e.g., morphine, fentanyl) for the relief of pain (e.g., cancer, back problems, HIV, post-surgery), local and systemic delivery of chemotherapy for the treatment of cancer (e.g., hepatic artery 5-FU infusion for liver tumors), medications for the treatment of cardiac conditions (e.g., anti-arrhythmic drugs for cardiac rhythm abnormalities), intrathecal delivery of anti-spasmotic drugs (e.g., baclofen) for spasticity in neurological disorders (e.g., Multiple Sclerosis, spinal cord injuries, brain injury, cerebral palsy), or local/regional antibiotics for infection management (e.g., osteomyelitis, septic arthritis).
  • narcotics e.g., morphine, fentanyl
  • chemotherapy e.g.
  • drug delivery pumps are implanted subcutaneously and consist of a pump unit with a drug reservoir and a flexible catheter through which the drug is delivered to the target tissue.
  • the pump stores and releases prescribed amounts of medication via the catheter to achieve therapeutic drug levels either locally or systemically (depending upon the application).
  • the center of the pump has a self-sealing access port covered by a septum such that a needle can be inserted percutaneously (through both the skin and the septum) to refill the pump with medication as required.
  • Constant-rate pumps are usually powered by gas and are designed to dispense drugs under pressure as a continual dosage at a preprogrammed, constant rate.
  • Programmable- rate pumps utilize a battery-powered pump and a constant pressure reservoir to deliver drugs on a periodic basis in a manner that can be programmed by the physician or the patient.
  • the drug may be delivered in small, discrete doses based on a programmed regimen which can be altered according to an individual's clinical response.
  • Implantable drug delivery pumps are implanted to deliver drug at a regulated dose and may, in certain applications, be used in conjunction with implantable sensors that collect information which is used to regulate drug delivery (often called a "closed loop" system).
  • Implantable drug delivery pumps may function and deliver drug in a variety of ways, which include, but are not limited to: (a) delivering drugs only when changes in the body are detected (e.g., sensor stimulated); (b) delivering drugs as a continuous slow release (e.g., constant flow); (c) delivering drugs at prescribed dosages in a pulsatile manner (e.g., non-constant flow); (d) delivering drugs by programmable means; and (e) delivering drugs through a device that is designed for a specific anatomical site (e.g., intraocular, intrathecal, intraperitoneal, intra-arterial or intracardiac).
  • a specific anatomical site e.g., intraocular, intrathecal, intraperitoneal, intra-art
  • drug delivery pumps may also be categorized based on their mechanical delivery technology (e.g., the driving force by which drug delivery occurs).
  • the mechanics for delivering drugs may include, without limitation, osmotic pumps, metering systems, peristaltic (roller) pumps, electronically driven pumps, ocular drug delivery pumps and implants, elastomeric pumps, spring-contraction pumps, gas-driven pumps (e.g., induced by electrolytic cell or chemical reaction), hydraulic pumps, piston-dependent pumps and non-piston-dependent pumps, dispensing chambers, infusion pumps, passive pumps, infusate pumps and osmotically-driven fluid dispensers.
  • an implantable drug delivery device or pump depends upon the device, particularly the catheter or drug-dispensing component(s), being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid) and not becoming encapsulated or obstructed by scar tissue.
  • target tissue e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid
  • these devices are implanted in the body, they are subject to a "foreign body" response from the surrounding host tissues as described previously.
  • the drug-delivery catheter lumen, catheter tip, dispensing components, or delivery membrane may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely.
  • the entire pump, the catheter and/or the dispensing components can become encapsulated by scar (i.e., the body "walls off' the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule).
  • scar i.e., the body "walls off' the device with fibrous tissue
  • the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule).
  • Either of these developments may lead to inefficient or incomplete drug flow to the desired target tissues or organs (and loss of clinical benefit), while encapsulation can also lead to local drug accumulation (in the capsule) and additional clinical complications (e.g., local drug toxicity; drug sequestration followed by sudden "dumping" of large amounts of drug into the surrounding tissues).
  • tissue surrounding the implantable pump can be inadvertently damaged from the inflammatory foreign body response leading to loss of function and/or tissue damage (e.g., scar tissue in the spinal canal causing pain or obstructing the flow of cerebrospinal fluid).
  • tissue damage e.g., scar tissue in the spinal canal causing pain or obstructing the flow of cerebrospinal fluid.
  • Implantable drug delivery pumps that release one or more therapeutic agents for reducing scarring at the device-tissue interface (particularly in and around the drug delivery catheter or drug dispensing components) may help prolong the clinical performance of these devices. Inhibition of fibrosis can make sure that the correct amount of drug is dispensed from the device at the appropriate rate and that potentially toxic drugs do not become sequestered in a fibrous capsule. For devices that include electrical or battery components, not only can fibrosis cause the device to function suboptimally or not at all, it can cause excessive drain on battery life as increased energy is required to overcome the increased resistance imposed by the intervening scar tissue.
  • the drug delivery pump may deliver drugs in a continuous, constant-flow, slow release manner.
  • the drug delivery pump may be a passive pump adapted to provide a constant flow of medication which may be regulated by a pressure sensing chamber and a valve chamber in which the constant flow rate may be changed to a new constant flow rate. See, e.g., U.S. Patent No. 6,589,205.
  • the drug delivery pump may deliver drugs at prescribed dosages in a non-constant flow or pulsatile manner.
  • the drug delivery pump may adapt a regular pump to generate a pulsatile fluid drug flow by continuously filling a chamber and then releasing a valve to provide a bolus pulse of the drug.
  • the drug delivery pump may be programmed to dispense drug in a very specific manner.
  • the drug delivery pump may be a programmable infusate pump composed of a variable volume infusate chamber, and variable volume control fluid pressure and displacement reservoirs, whereby a fluid flow is sampled by a microprocessor based on the programmed value and adjustments are made accordingly to maintain the programmed fluid flow. See, e.g., U.S. Patent No. 4,443,218.
  • the drug delivery pump suitable for use in the present invention may be manufactured based on different mechanical technologies (e.g., driving forces) of delivering drugs.
  • the drug delivery pump may be an implant composed of a piston that divides two chambers in which one chamber contains a water-swellable agent and the other chamber contains a leuprolide formulation for delivery. See, e.g., U.S. Patent No. 5,728,396.
  • the drug delivery pump may be a non-cylindrical osmotic pump system that may not rely upon a piston to infuse drug and conforms to the anatomical implant site. See, e.g., U.S. Patent No. 6,464,688.
  • the drug delivery pump may be an osmotically driven fluid dispenser composed of a flexible inner bag that contains the drug composition and a port in which the composition can be delivered. See, e.g., U.S. Patent No. 3,987,790.
  • the drug delivery pump may be a fluid-imbibing delivery implant composed of a compartment with a composition permeable to the passage of fluid and has an extended rigid sleeve to resist transient mechanical forces. See, e.g., U.S.
  • the drug delivery pump may be a pump with an isolated hydraulic reservoir, metering device, displacement reservoir, drug reservoir, and drug infusion port that is all contained in a housing apparatus. See, e.g., U.S. Patent No. 6,629,954.
  • the drug delivery pump may be composed of a dispensing chamber that has a dispensing passage and valves that are under compressive force to enable drug to flow in a one-way direction. See, e.g., U.S. Patent No. 6,283,949.
  • the drug delivery pump may be spring-driven based on a spring regulating pressure difference with a variable volume drug chamber. See, e.g., U.S. Patent No. 4,772,263.
  • osmotically driven drug delivery pumps that are commercially available and suitable for the practice of the invention.
  • These osmotic pumps include the DUROS Implant and ALZET Osmotic Pump from Alza Corporation (Mountain View, CA), which are used to delivery a wide variety of drugs and other therapeutics through the method of osmosis (see, e.g., U.S. Patent Nos. 6,283,953; 6,270,787; 5,660,847; 5,112,614; 5,030,216 and 4,976,966).
  • the drug delivery pump can be combined with an agent that inhibits fibrosis to improve performance of the device.
  • Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the device (e.g., the polymers that make up the delivery catheters, the semipermeable membranes etc.).
  • the fibrosis-inhibiting agent can be infiltrated into the region around the device-tissue interface. It may be obvious to one of skill in the art that commercial drug delivery pumps not specifically cited as well as next- generation and/or subsequently-developed commercial drug delivery products are to be anticipated and are suitable for use under the present invention.
  • the drug delivery pump may be an insulin pump.
  • Insulin pumps are used for patients with diabetes to replace the need to control blood glucose levels by daily manual injections of insulin. Precise titration of the dosage and timing of insulin administration is a critical component in the effective management of diabetes. If the insulin dosage is too high, blood glucose levels drop precipitously, resulting in confusion and potentially even loss of consciousness. If insulin dosage is too low, blood glucose levels rise too high, leading to excessive thirst, urination, and changes in metabolism known as ketoacidosis. If the timing of insulin administration is incorrect, blood glucose levels can fluctuate wildly between the two extremes - a situation that is thought to contribute to some of the long-term complications of diabetes such as heart disease, kidney failure, nerve damage and blindness. Since in the extreme, all these conditions can be life threatening, the precise dosing and timing of insulin administration is essential to preventing the short and long-term complications of diabetes.
  • Implantable pumps automate the administration of insulin and eliminate human errors of dosage and timing that can have long-term health consequences.
  • the pump has the capability to inject insulin regularly, multiple times a day and in small doses into the blood stream, peritoneal cavity or subcutaneous tissue.
  • the pump is refilled with insulin once or twice a month by injection directly into the pump chamber. This reduces the number of externally administered injections the patient must undergo and also allows preprogrammed variable amounts of insulin to be released at different times into the blood stream; a situation which more closely resembles normal pancreas function and minimizes fluctuations in blood glucose levels.
  • the insulin pump may be activated by an externally generated signal after the patient has withdrawn a drop of blood, subjected it to an analysis, and made a determination of the amount of insulin that needs to be delivered.
  • an externally generated signal after the patient has withdrawn a drop of blood, subjected it to an analysis, and made a determination of the amount of insulin that needs to be delivered.
  • this technology is the production of a closed- loop "artificial pancreas" which can continuously detect blood glucose levels (through an implanted sensor) and provide feedback to an implantable pump to modulate the administration of insulin to a diabetic patient.
  • the drug delivery pump may include both an implantable sensor and a drug delivery pump by being composed of a mass of living cells and an electrical signal that regulates the delivery of glucose or glucagon or insulin.
  • the drug delivery pump may be composed of a single channel catheter with a sensor which is implanted in a vessel that transmits blood chemistry to a subcutaneously implanted infusion device which then dispenses medication through the catheter. See, e.g., U.S. Patent No. 5,109,850.
  • the MINIMED 2007 Implantable Insulin Pump System from Medtronic MiniMed, Inc. (Northridge, CA).
  • the MINIMED pump delivers insulin into the peritoneal cavity in short, frequent bursts to provide insulin to the body similar to that of the normal pancreas (see, e.g., U.S. Patent Nos. 6,558,345 and 6,461 ,331).
  • the MINIMED 2001 Implantable Insulin Pump System (Medtronic MiniMed Inc., Northridge, CA) delivers intraperitoneal insulin injections in a pulsatile manner from a negative pressure reservoir. Both these devices feature a long catheter that transports insulin from the subcutaneously implanted pump into the peritoneal cavity.
  • the peritoneal drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely.
  • the insulin delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent.
  • Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters. Alternatively, or in addition, the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface.
  • intrathecal drug delivery pumps combined with a fibrosis-inhibitor can be used to may used to deliver drugs into the spinal cord for pain management and movement disorders.
  • Chronic pain is one of the most important clinical problems in all of medicine. For example, it is estimated that over 5 million people in the United States are disabled by back pain. The economic cost of chronic back pain is enormous, resulting in over 100 million lost work days annually at an estimated cost of $50-100 billion. The cost of managing pain for oncology patients is thought to approach $12 billion. Chronic pain disables more people than cancer or heart disease and costs the American public more than both cancer and heart disease combined. In addition to the physical consequences, chronic pain has numerous other costs including loss of employment, marital discord, depression and prescription drug addiction. It goes without saying, therefore, that reducing the morbidity and costs associated with persistent pain remains a significant challenge for the healthcare system.
  • Intractable severe pain resulting from injury, illness, scoliosis, spinal disc degeneration, spinal cord injury, malignancy, arachnoiditis, chronic disease, pain syndromes (e.g., failed back syndrome, complex regional pain syndrome) and other causes is a debilitating and common medical problem.
  • analgesics particularly drugs like narcotics, are not a viable solution due to tolerance, loss of effectiveness, and addiction potential.
  • intrathecal drug delivery devices have been developed to treat severe intractable back pain that is resistant to other traditional treatment modalities such as drug therapy, invasive therapy (surgery), or behavioral/lifestyle changes.
  • Intrathecal drug delivery pumps are designed and used to reduce pain by delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord. Typically, since this therapy delivers pain medication topically to pain receptors contained in the spinal cord that transmit pain sensation directly to the brain, smaller doses of medication are needed to gain relief. Morphine and other narcotics (usually fentanyl and sufentanil) are the most commonly delivered agents and many patients receive superior relief with lower doses than can be achieved with systemic delivery. Intrathecal drug delivery also allows the administration of pain medications (such as Ziconotide; an N-type calcium channel blocker made by Elan Pharmaceuticals) that cannot cross the blood-brain barrier and are thus only effective when administered by this route.
  • pain medications such as Ziconotide; an N-type calcium channel blocker made by Elan Pharmaceuticals
  • Intrathecal pumps are also used in the management of neurological and movement disorders.
  • Baclofen (marketed as Lioresal by Novartis) is an antispasmotic/muscle relaxant used to treat spasticity and improve mobility in patients with Multiple Sclerosis, cystic fibrosis and spinal injuries. This drug has been proven to be more effective and cause fewer side effects when administered into the CSF by an intrathecal drug delivery pump.
  • Efforts are also underway to treat epilepsy, brain tumors, Alzheimer's disease, Parkinson's disease and Amyetropic Lateral Sclerosis (ALS - Lou Gehrig's disease) via intrathecal administration of agents that may be too toxic to deliver systemically or do not cross the blood-brain barrier.
  • r- BDNF brain-derived neurotrophic factor
  • An intrathecal drug delivery system consists of an intrathecal drug infusion pump and an intraspinal catheter, both of which are fully implanted.
  • the pump device is implanted under the skin in the abdominal area, just above or below the beltline and can be refilled by percutaneous injection of the drug into the reservoir.
  • the catheter is tunneled under the skin and runs from the pump to the intrathecal space of the spine.
  • the pump administers prescribed amounts of medication to the cerebrospinal fluid in either a continuous fashion or in a manner than can be controlled by the physician or the patient in response to symptoms.
  • implantable intrathecal pumps are suitable for use in combination with a fibrosis-inhibiting agent in the practice of the invention.
  • the implantable pump used to deliver medication may be composed of two osmotic pumps with semipermeable membranes configured to deliver up to two drug delivery regimens at different rates, and having a built-in backup drug delivery system whereby the delivery of drug may continue when the primary delivery system reaches the end of its useful life or fails unexpectedly. See, e.g., U.S. Patent No. 6,471 ,688.
  • the implantable pump may be may be composed of a battery-operated pump unit with a drug reservoir, catheter, and electrodes that are implanted in the epidural space of a patient for relief of pain by delivering a liquid pain-relieving agent through the catheter to the desired location. See, e.g., U.S. Patent No. 5,458,631.
  • Implantable pumps may be implanted abdominally which then dispenses drug through a catheter that is tunneled from the abdominal implant site, through the neck to an entry site in the head, and then to the localized treatment site within the brain.
  • Pumps that deliver drug to the brain may discharge the drug at a variety of locations, including, but not limited to, anterior thalamus, ventrolateral thalamus, internal segment of the globus pallidus, substantia nigra pars reticulate, subthalamic nucleus, external segment of globus pallidus, and neostriatum.
  • the drug delivery pump may be composed of an implantable pump portion coupled to a catheter for infusing dosages of drug to a predetermined location of the brain when a sensor detects a symptom, such that a neurological disorder (e.g., seizure) may be treated. See, e.g., U.S. Patent No. 5,978,702.
  • the implantable pump may be implanted adjacent to a predetermined infusion site in a brain such that a predetermined dosage of at least one drug capable of altering the level of excitation of neurons of the brain may be infused such that neurodegeneration is prevented and/or treated.
  • the implantable pump may include a reservoir for the therapeutic agent which is stored between the galea aponeurotica and cranium of a subject whereby drug is then dispensed via pumping action to the desired location. See, e.g., U.S. Patent No. 6,726,678.
  • the SYNCHROMED EL Infusion System which is made by Medtronic, Inc. and is indicated for chronic Intrathecal Baclofen Therapy (ITB Therapy) (see, e.g., U.S. Patent Nos. 6,743,204; 6,669,663; 6,635,048; 6,629,954; 6,626,867; 6,102,678; 5,978,702 and 5,820,589)
  • the SYNCHROMED pump is a programmable, battery-operated device that stores and delivers medication based on the programmed dosing regimen. Medtronic, Inc.
  • All these devices feature a long catheter that transports the active agent from a subcutaneously implanted pump into the intrathecal space in the spinal cord.
  • the intrathecal drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely.
  • Another potential complication with intrathecal drug delivery is the formation of fibrous tissue in the subdural space that can obstruct CSF flow and lead to serious complications (e.g., hydrocephalus, increased intracranial pressure).
  • the drug delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent and/or prevents fibrosis in the surrounding tissue.
  • Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters. Alternatively, or in addition, the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface.
  • the adjuvant use of an anti-infective agent as a catheter coating and /or implant, with or without a fibrosis-inhibiting agent, may also be beneficial in the practice of this invention.
  • the drug delivery pump may be a pump that dispenses a chemotherapeutic drug for the treatment of cancer.
  • Pumps for dispensing a drug for the treatment of cancer are used to deliver chemotherapeutic agents to a local area of the body.
  • FUDR 2'-deoxy 5-fluorouridine
  • adenocarcinoma colon, breast, stomach
  • FUDR 2'-deoxy 5-fluorouridine
  • the drug is delivered via an implantable pump into the artery which provides blood supply to the liver. This allows for higher drug concentrations to reach the liver (the drug is not diluted in the blood as may occur in intravenous administration) and prevents clearance by the liver (the drug is metabolized by the liver and may be rapidly cleared from the bloodstream if administered i.v.); both of which allow higher concentrations of the drug to reach the tumor.
  • the implantable pump may have a dispensing chamber with a dispensing passage and actuator, reservoir housing with reservoir, and septum for refilling the reservoir.
  • the implantable pump may have a dispensing chamber with a dispensing passage and actuator, reservoir housing with reservoir, and septum for refilling the reservoir.
  • Medtronic, Inc. sells their ISOMED Constant-Flow Infusion System which may be used to deliver chronic intravascular infusion of floxuridine in a fixed flow rate for the treatment of primary or metastatic cancer.
  • Tricumed Medizintechnik GmbH sells their ARCHIMEDES DC implantable infusion pump specially adapted to deliver chemotherapy in a constant flow rate within the vicinity of a tumor (see, e.g., U.S. Patent Nos. 5,908,414 and 5,769,823).
  • Arrow International produces the Model 3000 infusion pump that provides constant-rate administration of chemotherapeutic agents into a tumor. All these devices feature a catheter that transports the chemotherapeutic agent from a subcutaneously implanted pump directly into the tumor or the artery that supplies a tumor.
  • the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely.
  • the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by neointimal tissue which may impair the flow of drug into the blood vessel.
  • the drug delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters. Alternatively, or in addition, the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface.
  • an anti-infective agent as a catheter coating and /or implant, with or without a fibrosis-inhibiting agent, may also be beneficial in the practice of this invention. It may be obvious to one of skill in the art that commercial chemotherapy delivery pumps and implants not specifically cited as well as next-generation and/or subsequently-developed commercial chemotherapy delivery products are to be anticipated and are suitable for use in the present invention.
  • the drug delivery pump may be a pump that dispenses a drug for the treatment of heart disease.
  • Pumps for dispensing a drug for the treatment of heart disease may be used to treat conditions including, but not limited to atrial fibrillation and other cardiac rhythm disorders.
  • Atrial fibrillation is a form of heart disease that afflicts millions of people. It is a condition in which the normal coordinated contraction of the heart is disrupted, primarily by abnormal and uncontrolled action of the atria of the heart. Normally, contractions occur in a controlled sequence with the contractions of the other chambers of the heart. When the right atrium fails to contract, contracts out of sequence, or contracts ineffectively, blood flow from the atria to the ventricles is disrupted.
  • Atrial fibrillation can cause weakness, shortness of breath, angina, lightheadedness and other symptoms due to reduced ventricular filling and reduced cardiac output. Stroke can occur as a result of clot forming in a poorly contracting atria, breaking loose, and traveling via the bloodstream to the arteries of the brain where they become wedged and obstruct blood flow (which may lead to brain damage and death).
  • atrial fibrillation is treated by medical or electrical conversion (defibrillation), however, complications may exist whereby the therapy causes substantial pain or has the potential to initiate a life threatening ventricular arrhythmia. The pain associated with the electrical shock is severe and unacceptable for many patients, since they are conscious and alert when the device delivers electrical therapy.
  • Medical therapy involves the delivery of anti-arrhythmic drugs by injecting them intravenously, administering them orally or delivering them locally via a drug delivery pump.
  • implantable pumps Numerous types of implantable pumps are described for dispensing a drug for the treatment of heart disease and are suitable for use in the practice of the invention.
  • the drug delivery pump may be an implantable cardiac electrode which delivers stimulation energy and dispenses drug adjacent to the stimulation site. See, e.g., U.S. Patent No. 5,496,360.
  • the drug delivery pump may have a plurality of silicone septii to facilitate the filling of drug reservoirs within the pump which is subcutaneously implanted with a catheter which travels transvenously by way of the subclavian vein through the superior vena cava and into the right atrium for drug delivery. See, e.g., U.S. Patent No. 6,296,630.
  • the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. If placed intravascularly, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by neointimal tissue which may impair the flow of drug into the blood vessel or the right atrium.
  • the drug delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent.
  • Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters.
  • the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface.
  • the adjuvant use of an anti-infective agent as a catheter coating and /or implant, with or without a fibrosis-inhibiting agent, may also be beneficial in the practice of this invention.
  • Debiotech S.A. (Switzerland) has developed the MIP device which is an implantable piezo-actuated silicon micropump for programmable drug delivery applications.
  • This high-performance micropump is based on a MEMS (Micro-Electro-Mechanical) system which allows it to . maintain a low flow rate.
  • the DUROS sufentanil implant from Durect Corporation (Cupertino, CA) is a titanium cylinder that contains a drug reservoir, and a piston driven by an osmotic engine.
  • the VIADUR (leuprolide acetate) implant available from Alza Corporation uses the same DUROS implant technology to deliver leuprolide over a 12 month period to reduces testosterone levels for the treatment prostate cancer (see, e.g., U.S. Patent Nos. 6,283,953; 6,270,787; 5,660,847; 5,112,614; 5,030,216 and 4,976,966).
  • Fibrous encapsulation of the device can cause failure in a number of ways including: obstructing the semipermeable membrane (which will impair functioning of the osmotic engine by preventing the flow of fluids into the engine), obstructing the exit port (which will impair drug flow out of the device) and/or complete encapsulation (which will create a microenvironment that prevents drug distribution).
  • the drug delivery implant can be combined with an agent that inhibits fibrosis to prevent encapsulation, prevent obstruction of the semipermeable membrane and/or to keep the delivery port patent. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the drug delivery implant. Alternatively, or in addition, the fibrosis- inhibiting agent may be infiltrated into the tissue around the drug delivery implant.
  • an implantable drug delivery device or pump depends upon the device, particularly the catheter or drug-dispensing component(s), being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid) and not becoming encapsulated or obstructed by scar tissue.
  • target tissue e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid
  • the drug- delivery catheter lumen, catheter tip, dispensing components, or delivery membrane may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely.
  • the entire pump, the catheter and/or the dispensing components can become encapsulated by scar (i.e., the body "walls off' the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule).
  • scar i.e., the body "walls off' the device with fibrous tissue
  • the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule).
  • Either of these developments may lead to inefficient or incomplete drug flow to the desired target tissues or organs (and loss of clinical benefit), while encapsulation can also lead to local drug accumulation (in the capsule) and additional clinical complications (e.g., local drug toxicity; drug sequestration followed by sudden "dumping" of large amounts of drug into the surrounding tissues).
  • Implantable pumps that include electrical or battery components, not only can fibrosis cause the device to function suboptimally or not at all, it can cause excessive drain on battery life as increased energy is required to overcome the increased resistance imposed by the intervening scar tissue.
  • Implantable pumps that release a therapeutic agent for reducing scarring at the device-tissue interface can be used to increase efficacy, prolong clinical performance, ensure that the correct amount of drug is dispensed from the device at the appropriate rate, and reduce the risk that potentially toxic drugs become sequestered in a fibrous capsule.
  • the present invention provides implantable pumps that include a fibrosis-inhibiting agent or a composition that includes a fibrosis-inhibiting agent. Numerous polymeric and non-polymeric delivery systems for use in implantable pumps have been described above. These compositions can further include one or more fibrosis- inhibiting agents such that the overgrowth of granulation or fibrous tissue is inhibited or reduced.
  • Methods for incorporating fibrosis-inhibiting compositions onto or into implantable drug delivery pumps to reduce scarring at the device-tissue interface include: (a) directly affixing to the implantable pump, catheter and/or drug dispensing components a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier), (b) directly incorporating into the implantable pump, catheter and/or drug dispensing components a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier (c) by coating the implantable pump, catheter and/or drug dispensing components with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the implantable pump,
  • the coating process can be performed in such a manner as to: (a) coat a portion of the device (such as the catheter, drug delivery port, semipermeable membrane); or (b) coat the entire device with the fibrosis-inhibiting composition.
  • the fibrosis- inhibiting agent can be mixed with the materials that are used to make the implantable pump such that the fibrosis-inhibiting agent is incorporated into the final product.
  • a medical device may be prepared which has a coating, where the coating is, e.g., uniform, non-uniform, continuous, discontinuous, or patterned.
  • an implantable drug delivery pump device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug (i.e., one or more fibrosis-inhibiting agents).
  • the reservoirs may be formed from divets in the device surface or micropores or channels in the device body.
  • the reservoirs are formed from voids in the structure of the device.
  • the reservoirs may house a single type of drug (e.g., fibrosis-inhibiting agent) or more than one type of drug (e.g., a fibrosis-inhibiting agent and an anti-infective agent).
  • the drug(s) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs.
  • a carrier e.g., a polymeric or non-polymeric material
  • the filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier.
  • the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount (dose) of drug, and each layer may have a different composition to further tailor the amount and type of drug that is released from the substrate.
  • the multi-layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void.
  • the coating of the medical device may directly contact the pump, or it may indirectly contact the pump when there is something, e.g., a polymer layer, that is interposed between the pump and the
  • the fibrosis-inhibiting agent can be applied directly or indirectly to the tissue adjacent to the implantable pump (preferably near in the tissue adjacent to where the drug is delivered from the device).
  • the fibrosis-inhibiting agent with or without a polymeric, non-polymeric, or secondary carrier: (a) to the implantable pump, catheter and/or drug dispensing component surface (e.g., as an injectable, paste, gel, or mesh) during the implantation procedure; (b) to the surface of the tissue ⁇ e.g., as an injectable, paste, gel, in situ forming gel, or mesh) prior to, immediately prior to, or during, implantation of the implantable pump, catheter and/or drug dispensing components; (c) to the surface of the implantable pump, catheter and/or drug dispensing components and/or to the tissue surrounding the implanted pump, catheter and/or drug dispensing components (e.g., as an injectable, paste, gel, in situ forming gel, or mesh) immediately after implantation; (d) by topical application of the anti-fibrosis agent into the anatomical space where the implantable pump, catheter and/or drug dispensing components will be placed (particularly useful for
  • polymeric carriers themselves can help prevent the formation of fibrous tissue around the implanted pump, catheter and/or drug dispensing components. These carriers (described below) are particularly useful for the practice of this embodiment, either alone, or in combination with a fibrosis-inhibiting composition.
  • polymeric carriers can be infiltrated (as described in the previous paragraph) into the vicinity of the interface between the implanted pump, catheter and/or drug dispensing components of the device and the tissue and include: (a) sprayable collagen-containing formulations such as COSTASIS and CT3, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (b) sprayable PEG-containing formulations such as COSEAL, FOCALSEAL , SPRAYGEL or DURASEAL, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (d) hyaluronic acid
  • a preferred polymeric matrix which can be used to help prevent the formation of fibrous tissue around the implanted pump, catheter and/or drug dispensing components, either alone or in combination with a fibrosis inhibiting agent/composition is formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents.
  • reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-s
  • Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents.
  • Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500.
  • collagen or a collagen derivative is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue around the implanted pump, catheter and/or drug dispensing components.
  • collagen or a collagen derivative e.g., methylated collagen
  • any anti-scarring agent described below may be utilized alone, or in combination, in the practice of this embodiment.
  • implantable pumps and their drug delivery mechanisms e.g., catheters, ports etc.
  • Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered can be measured, and appropriate surface concentrations of active drug can be determined.
  • the fibrosis-inhibiting agents used alone or in combination, may be administered under the following dosing guidelines:
  • Therapeutic agents that may be used include but are not limited to ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglon
  • Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 .
  • mTOR inhibitors including AP- 23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 . Minimum concentration of 10 ⁇ - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • Minimum concentration of 10 ⁇ - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • TNF alpha antagonists including Etanercept, Humicade, Adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg.
  • Minimum concentration of 10 " 8 - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg.
  • NF Kappa B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg.
  • Minimum concentration of 10 " 8 - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • K Elongation Factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 ⁇ g to 1000 mg); preferred 1 ⁇ g to 500 mg.
  • anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC 50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus.
  • selected assays described herein such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus.
  • the total dose typically should not exceed 200 mg (range of 0.1 ⁇ g to 200 mg) and preferably 1 ⁇ g to 100 mg; dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 ; and minimum concentration of 10 '8 - 10 "4 M of agent should be maintained on the implant or barrier surface.
  • agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC 50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate.
  • the total dose typically should not to exceed 500 mg (range of 1.0 ⁇ g to 500 mg) and preferably 1 ⁇ g to 200 mg; dose per unit area of 0.01 ⁇ g - 200 ⁇ g per mm 2 , preferably 0.1 ⁇ g/mm 2 - 40 ⁇ g/mm 2 ; and minimum concentration of 10 '8 - 10 "4 M of agent should be maintained on the implant or barrier surface.
  • agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
  • the total dose typically should not exceed 1000 mg (range of 0.1 ⁇ g to 1000 mg), preferably 1 ⁇ g to 500 mg; dose per unit area of 0.01 ⁇ g - 500 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 100 ⁇ g/mm 2 ; and minimum concentration of 10 '8 - 10 ⁇ 4 M of agent should to be maintained on the implant or barrier surface.
  • 1000 mg range of 0.1 ⁇ g to 1000 mg
  • dose per unit area 0.01 ⁇ g - 500 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 100 ⁇ g/mm 2 ; and minimum concentration of 10 '8 - 10 ⁇ 4 M of agent should to be maintained on the implant or barrier surface.
  • the invention provides for medical devices that include an agent that inhibits this tissue accumulation in the vicinity of the device, i.e., between the medical device and the host into which the medical device is implanted.
  • the agent is therefore effective for this goal, is present in an amount that is effective to achieve this goal, and is present at one or more locations that allow for this goal to be achieved, and the device is designed to allow the beneficial effects of the agent to occur.
  • these therapeutic agents can be used alone, or in combination, to prevent scar tissue build-up in the vicinity of the device-tissue interface in order to improve the clinical performance and longevity of these implants.
  • Suitable therapeutic agents for use in the invention may be readily identified based upon in vitro and in vivo (animal) models, such as those provided in Examples 34-44, 46, and 54. Agents which inhibit fibrosis can also be identified through in vivo models including inhibition of intimal hyperplasia development in the rat balloon carotid artery model (Examples 37 and 46). The assays set forth in Examples 36 and 44 may be used to determine whether an agent is able to inhibit cell proliferation in fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC 50 for inhibition of cell proliferation within a range of about 10 "6 to about 10 "10 M.
  • the agent may have an IC 50 for inhibition of cell proliferation of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 40 may be used to determine whether an agent may inhibit migration of fibroblasts and/or smooth muscle cells.
  • the agent has an IC 50 for inhibition of cell migration within a range of about 10 "6 to about 10 '9 M.
  • the agent may have an IC 50 for inhibition of fibroblast or smooth muscle cell migration of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • Assays set forth herein may be used to determine whether an agent is able to inhibit inflammatory processes, including nitric oxide production in macrophages (Example 34), and/or TN F-alpha production by macrophages (Example 35), and/or IL-1 beta production by macrophages (Example 41), and/or IL-8 production by macrophages (Example 42), and/or inhibition of MCP- 1 by macrophages (Example 43).
  • the agent has an IC 50 for inhibition of any one of these inflammatory processes within a range of about 10 "6 to about 10 '10 M.
  • the agent may have an IC 50 for any one of these inflammatory processes of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 38 may be used to determine whether an agent is able to inhibit MMP production.
  • the agent has an IC 50 for inhibition of MMP production within a range of about 10 "4 to about 10 "8 M. .
  • the agent may have an IC 50 for inhibition of MMP production of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 41 (also known as the CAM assay) may be used to determine whether an agent is able to inhibit angiogenesis.
  • the agent has an IC 50 for inhibition of angiogenesis within a range of about 10 '6 to about 10 '10 M. In certain embodiments, the agent may have an IC 50 for inhibition of angiogenesis of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 38 may be used to determine whether an agent is able to inhibit MMP- 1.
  • the agent has an IC 50 for inhibition of MMP-1 within a range of about 10 " ⁇ to about 10 "10 M.
  • the agent may have an IC 5O for inhibition of MMP-1 of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • an agent which is useful for the compositions and methods described herein may exhibit different levels of effectiveness in different assays (e.g., effectiveness indicated by IC 50 values) and in different animal models described herein and practiced in the art.
  • an agent that is useful may exhibit activity in one assay or animal model or may exhibit activity in more than one, two, or three, etc., different assays and/or animal models.
  • Agents which reduce the formation of surgical adhesions may be identified through in vivo models including, for example, the rabbit surgical adhesions model and the rat caecal sidewall model. These pharmacologically active agents (described below) can then be delivered at appropriate dosages into to the tissue either alone, or via carriers (described herein), to treat the clinical problems described herein.
  • the fibrosis-inhibiting compound is an adensosine A2A receptor antagonist (e.g., Sch-63390 (Schering-Plough) or an A2A receptor antagonists from Almirall-Prodesfarma, SCH-58261 (CAS No. 160098-96-4), or an analogue or derivative thereof).
  • an adensosine A2A receptor antagonist e.g., Sch-63390 (Schering-Plough) or an A2A receptor antagonists from Almirall-Prodesfarma, SCH-58261 (CAS No. 160098-96-4), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an AKT inhibitor (e.g., PKB inhibitors from DeveloGen, AKT inhibitors from Array BioPharma, Celgene, Merck & Co, Amphora, NeoGenesis Pharmaceuticals, A- 443654 (Abbott Laboratories), erucylphosphocholine (AEterna Zentaris), KRX- 401 (Keryx), protein kinase B inhibitors from Astex Technology, PX-316 (ProlX), or an analogue or derivative thereof).
  • AKT inhibitor e.g., PKB inhibitors from DeveloGen, AKT inhibitors from Array BioPharma, Celgene, Merck & Co, Amphora, NeoGenesis Pharmaceuticals, A- 443654 (Abbott Laboratories), erucylphosphocholine (AEterna Zentaris), KRX- 401 (Keryx), protein kinase B inhibitors from Astex Technology, PX-316 (ProlX
  • the fibrosis-inhibiting compound is an alpha 2 integrin antagonist (e.g., Pharmaprojects No. 5754 (Merck KGaA), or an analogue or derivative thereof). 4.
  • Alpha 4 Integrin Antagonist e.g., Pharmaprojects No. 5754 (Merck KGaA), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an alpha 4 integrin antagonist (e.g., T-0047 (Tanabe Seiyaku), VLA-4 antagonists from Sanofi-Aventis, Merck & Co., Biogen personal, Uriach, and Molecumetics, alpha 4 integrin antagonists from Genentech), BIO-2421 (Biogen personal), cell adhesion inhibitors from Kaken Pharmaceuticals, CT-737 (Wyeth), CT-767 (Elan), CY-9652 (Epimmune), CY-9701 (Epimmune), fibronectin antagonists from Uriach, integrin alpha4 ⁇ 7 antagonists frin Wilex, Pharmaprojects No.
  • alpha 4 integrin antagonist e.g., T-0047 (Tanabe Seiyaku), VLA-4 antagonists from Sanofi-Aventis, Merck & Co., Biogen personal, and Molecumetics, alpha 4 integrin antagonists from Genentech), BIO
  • the fibrosis-inhibiting compound is an alpha 7 nicotinic receptor agonist (e.g., AZD-0328 (AstraZeneca), galantamine (CAS No. 357-70-0) (Synaptc), MEM-3454 or nicotinic alpha-7 agonist (Memory Pharmaceuticals and Critical Therapeutics), Pharmaprojects No. 4779 (AstraZeneca), PNU-282987 (Pfizer), SSR-180711 (Sanofi-Aventis), TC-1698 or TC-5280 (Targacept), or an analogue or derivative thereof).
  • alpha 7 nicotinic receptor agonist e.g., AZD-0328 (AstraZeneca), galantamine (CAS No. 357-70-0) (Synaptc), MEM-3454 or nicotinic alpha-7 agonist (Memory Pharmaceuticals and Critical Therapeutics), Pharmaprojects No. 4779 (AstraZeneca),
  • the fibrosis-inhibiting compound is an angiogenesis inhibitor (e.g., AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (No
  • the fibrosis-inhibiting compound is an apoptosis antagonist (e.g., didemnin B, RGB-286199 (GPC Biotech), 5F-DF- 203 (Cancer Research Technology), aplidine, bongkrekic acid, triammonium salt, [6]-gingerol (CAS No. 23513-14-6), or an analogue or derivative thereof).
  • apoptosis antagonist e.g., didemnin B, RGB-286199 (GPC Biotech), 5F-DF- 203 (Cancer Research Technology), aplidine, bongkrekic acid, triammonium salt, [6]-gingerol (CAS No. 23513-14-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an apoptosis activator (e.g., aplidine (CAS No. 137219-37-5) (PharmaMar), canfosfamide hydrochloride (CAS No. 58382-37-74 and 39943-59-6) (Telik), idronoxil (CAS No. 81267-65-4) (Novogen), OSI-461 (OSI Pharmaceuticals), DE-098 (Santen), ARQ-550RP (ArQuIe), ABJ-879 (Novartis), adaphostin (NIH), anticancer agents from Apogenix Biotechnology and.
  • apoptosis activator e.g., aplidine (CAS No. 137219-37-5) (PharmaMar), canfosfamide hydrochloride (CAS No. 58382-37-74 and 39943-59-6) (Telik), idronoxil (CAS No. 81267-65-4)
  • the fibrosis-inhibiting compound is a beta 1 integrin antagonist (e.g., ⁇ -1 integrin antagonists, Berkeley Lab, or an analogue or derivative thereof).
  • a beta 1 integrin antagonist e.g., ⁇ -1 integrin antagonists, Berkeley Lab, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a beta tubulin inhibitor (e.g., ZEN-017 (AEterna Zentaris), laulimalide (Kosan Biosciences), or an analogue or derivative thereof).
  • a beta tubulin inhibitor e.g., ZEN-017 (AEterna Zentaris), laulimalide (Kosan Biosciences), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an agent that blocks enzyme production in hepatitis C (e.g., merimepodib (Vertex Pharmaceuticals), or an analogue or derivative thereof).
  • hepatitis C e.g., merimepodib (Vertex Pharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a Bruton's tyrosine kinase inhibitor (e.g., a Btk inhibitor from Cellular Genomics, or an analogue or derivative thereof).
  • a Bruton's tyrosine kinase inhibitor e.g., a Btk inhibitor from Cellular Genomics, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a calcineurin inhibitor (e.g., tacrolimus (LifeCycle Pharma), or an analogue or derivative thereof).
  • a calcineurin inhibitor e.g., tacrolimus (LifeCycle Pharma)
  • analogue or derivative thereof e.g., tacrolimus (LifeCycle Pharma)
  • the fibrosis-inhibiting compound is a caspase 3 inhibitor (e.g., NM-3 (Mercian), or an analogue or derivative thereof).
  • a caspase 3 inhibitor e.g., NM-3 (Mercian), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a CC chemokine receptor antagonist (e.g., a chemokine receptor 3 antagonist, a chemokine receptor 6 antagonist, and a chemokine receptor 7 antagonist).
  • CC chemokine receptor antagonists include chemokine antagonists such as the CCR7 antagonists from Neurocrine Biosciences.
  • the fibrosis-inhibiting compound is a CC chemokine receptor antagonist (CCR) 1 , 3, & 5 (e.g., peptide T (Advanced lmmuni T), a CCR3 antagonist from GlaxoSmithKline, a chemokine antagonist (Pharmaprojects No. 6322) from Neurocrine Biosciences or Merck & Co., an HIV therapy agent from ReceptoPharm (Nutra Pharma), Pharmaprojects No. 6129 (Sangamo BioSciences), or an analogue or derivative thereof).
  • the CCCR antagonist is a CCR2b chemokine receptor antagonist such as RS 102895 (CAS No. 300815-41-2). 16.
  • the fibrosis-inhibiting compound is a cell cycle inhibitor (e.g., SNS-595 (Sunesis), homoharringtonine, or an analogue or derivative thereof).
  • the cell cycle inhibitor is an anti- microtubule agent (e.g., synthadotin, or an analogue or derivative thereof).
  • cell cycle inhibitor is a microtubule stimulant (e.g., KRX-0403, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a cathepsin B inhibitor (e.g., AM-4299A (Asahi Kasei Pharma), BDI-7800 (Biopharmacopae), a cathepsin B inhibitor from Axys (Celera Genomics), MDL- 104903 (CAS No. 180799-56-8) (Sanofi-Aventis), NC-700 (Nippon Chemiphar), Pharmaprojects No. 2332 (Hoffmann-La Roche), Pharmaprojects No. 4884 (Takeda), Pharmaprojects No. 5134 (Nippon Chemiphar), or an analogue or derivative thereof).
  • a cathepsin B inhibitor e.g., AM-4299A (Asahi Kasei Pharma), BDI-7800 (Biopharmacopae), a cathepsin B inhibitor from Axys (Celera Genomics), MDL- 104903 (CAS No. 180799-56-8) (
  • the fibrosis-inhibiting compound is a cathepsin K inhibitor (e.g., 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof).
  • a cathepsin K inhibitor e.g., 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cathepsin L Inhibitor (e.g., a cathepsin L inhibitor from Takeda, INPL-022-E10 (Amura Therapeutics), Pharmaprojects No. 5447 (Taiho), or an analogue or derivative thereof).
  • a cathepsin L Inhibitor e.g., a cathepsin L inhibitor from Takeda, INPL-022-E10 (Amura Therapeutics), Pharmaprojects No. 5447 (Taiho), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a CD40 antagonists ⁇ e.g., 5D12 (Chiron), ABI-793 (Novartis), an anticancer antibody from Chiron, anti-CD40 MAb-2 (Kirin Brewery), anti-CD40 (EIi Lilly), anti-CD40L antibody (UCB), a CD40 inhibitor from Apoxis, CD40 ligand inhibitor from Millennium Pharmaceuticals, a CD40/CAP inhibitor from Snow Brand, CGEN-40 (Compugen), CHIR-12.12 (Chiron), Pharmaprojects No.
  • CD40 antagonists ⁇ e.g., 5D12 (Chiron), ABI-793 (Novartis), an anticancer antibody from Chiron, anti-CD40 MAb-2 (Kirin Brewery), anti-CD40 (EIi Lilly), anti-CD40L antibody (UCB), a CD40 inhibitor from Apoxis, CD40 ligand inhibitor from Millennium Pharmaceuticals, a CD40/CAP inhibitor from Snow Brand, CGEN-40 (Com
  • the fibrosis-inhibiting compound is a chemokine receptor agonist (e.g., a chemokine agonist from NeuroTarget, or an analogue or derivative thereof).
  • a chemokine receptor agonist e.g., a chemokine agonist from NeuroTarget, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a chymase inhibitor (e.g., BL-3875 (Dainippon), LEX-043 (SuperGen), NK-3201 (CAS No. 204460-24-2) (Nippon Kayaku), or an analogue or derivative thereof).
  • a chymase inhibitor e.g., BL-3875 (Dainippon), LEX-043 (SuperGen), NK-3201 (CAS No. 204460-24-2) (Nippon Kayaku), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a collagenase (interstitial) antagonist (e.g., IBFB-212543 (IBFB Pharma), Pharmaprojects No. 3762 (Sanofi-Aventis), S-0885 (CAS No. 117517-22-3) (Sanofi-Aventis), SC-40827 (CAS No. 101470-42-2) (Pfizer), or an analogue or derivative thereof).
  • IBFB-212543 IBFB Pharma
  • Pharmaprojects No. 3762 Sanofi-Aventis
  • S-0885 CAS No. 117517-22-3
  • SC-40827 CAS No. 101470-42-2
  • the fibrosis-inhibiting compound is a CXCR (2, 4) antagonist (e.g., SB-656933 (GlaxoSmithKline), AMD3100 octahydrochloride (CAS No. 155148-31-5), or an analogue or derivative thereof).
  • CXCR (2, 4) antagonist e.g., SB-656933 (GlaxoSmithKline), AMD3100 octahydrochloride (CAS No. 155148-31-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cyclin dependent kinase (CDK) inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK-1 inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK-2 inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK- 4 inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK-6 inhibitor.
  • Representative examples of cyclin dependent kinase inhibitors include CAK1 inhibitors from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), or an analogue or derivative thereof.
  • Additional exemplary cyclin dependent protein kinase inhibitors include an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), CGP74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cyclooxygenase inhibitor (e.g., NS-398 (CAS No. 123653-11-2), ketoprofen, or an analogue or derivative thereof).
  • the cyclooxygenase inhibitor is a COX-1 inhibitor such as triflusal, or an analogue or derivative thereof).
  • DHFR Dihvdroorotate Dehydrogenase Inhibitor
  • the fibrosis-inhibiting compound is a DHFR inhibitor (e.g., PDX (Allos Therapeutics), SC12267, sulfamerazine (CAS No. 127-79-7), or an analogue or derivative thereof).
  • a DHFR inhibitor e.g., PDX (Allos Therapeutics), SC12267, sulfamerazine (CAS No. 127-79-7), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a dual integrin inhibitor (e.g., R411 (Roche Pharmaceuticals), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an elastase inhibitor (e.g., orazipone, depelestat (CAS No. 506433-25-6) (Dyax), AE-3763 (Dainippon), or an analogue or derivative thereof).
  • elastase inhibitor e.g., orazipone, depelestat (CAS No. 506433-25-6) (Dyax), AE-3763 (Dainippon), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an elongation factor-1 alpha inhibitor (e.g., aplidine, or an analogue or derivative thereof).
  • an elongation factor-1 alpha inhibitor e.g., aplidine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an endothelial growth factor (EGF) antagonist (e.g., neovastat, NM-3 (Mercian), or an analogue or derivative thereof).
  • EGF endothelial growth factor
  • the fibrosis-inhibiting compound is an endothelial growth factor receptor (EGF-R) kinase inhibitor (e.g., sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No.
  • EGF-R endothelial growth factor receptor
  • a KDR inhibitor from LG Life Sciences, CT-6685 or CT-6729 (UCB), KRN-633 or KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGF-R inhibitor such as SU 1498, a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an endothelial growth factor receptor 2 kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
  • an endothelial growth factor receptor 2 kinase inhibitor e.g., sorafenib tosylate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an endotoxin antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an epothilone or tubulin binder (e.g., ixabepilone (BMS), or an analogue or derivative thereof).
  • BMS ixabepilone
  • the fibrosis-inhibiting compound is an estrogen receptor antagonist (e.g., ERB-041 (Wyeth), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a FGF inhibitor (e.g., IDN-5390 (Indena), or an analogue or derivative thereof).
  • FGF inhibitor e.g., IDN-5390 (Indena), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an inhibitor of farnexyl transferase (FTI).
  • FTI farnexyl transferase
  • the FTI inhibits the RAS oncogene family.
  • FTI's include SARASAR (from Schering Corporation, Kenilworth, NJ), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a farnesyltransferase inhibitor (e.g., A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIlI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), or an analogue or derivative thereof).
  • a farnesyltransferase inhibitor e.g., A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIlI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a FLT- 3 kinase inhibitor (e.g. , Amphora, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a FGF receptor kinase inhibitor (e.g., MED-A300 (Gerolymatos), SSR-128129 (Sanofi- Aventis), TBC-2250 (Encysive Pharmaceuticals), XL-999 (Exelixis), or a FGF receptor kinase inhibitor from Paradigm Therapeutics, or an analogue or derivative thereof).
  • a FGF receptor kinase inhibitor e.g., MED-A300 (Gerolymatos), SSR-128129 (Sanofi- Aventis), TBC-2250 (Encysive Pharmaceuticals), XL-999 (Exelixis), or a FGF receptor kinase inhibitor from Paradigm Therapeutics, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a fibrinogen antagonist (e.g., AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, or an analogue or derivative thereof).
  • a fibrinogen antagonist e.g., AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (San
  • the fibrosis-inhibiting compound is a heat shock protein 90 antagonist (e.g., SRN-005 (Sirenade), geldanamycin or a derivative thereof, such as NSC-33050 (17-allylaminogeldanamycin; 17-AAG) or ⁇ -dimethylaminoethylamino- ⁇ -demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), radicicol, Humicola fuscoatra (CAS No. 12772-57-5), or an analogue or derivative thereof).
  • SRN-005 Serenade
  • geldanamycin or a derivative thereof such as NSC-33050 (17-allylaminogeldanamycin; 17-AAG) or ⁇ -dimethylaminoethylamino- ⁇ -
  • the fibrosis-inhibiting compound is a histone deacetylase inhibitor (e.g., FK228 (Gloucester), trichostatin A from Streptomyces sp. (CAS No. 58880-19-6), or an analogue or derivative thereof).
  • a histone deacetylase inhibitor e.g., FK228 (Gloucester), trichostatin A from Streptomyces sp. (CAS No. 58880-19-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an HMGCoA reductase inhibitor (e.g., an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na )CAS No. 143201-11-0), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an
  • ICAM inhibitor e.g., alicaforsen (CAS No. 185229-68-9) (ISIS Pharmaceuticals), an ICAM-5 modulator (such as ICAM-4 from ICOS), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an IL-
  • ICE & IRAK antagonist e.g., CJ-14877 or CP-424174 (Pfizer), NF-61 (Negma-Lerads), or an analogue or derivative thereof.
  • ICE & IRAK antagonist e.g., CJ-14877 or CP-424174 (Pfizer), NF-61 (Negma-Lerads), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an IL-2 inhibitor (e.g., AVE 8062 (Sanofi-Aventis), or an analogue or derivative thereof).
  • IL-2 inhibitor e.g., AVE 8062 (Sanofi-Aventis), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an immunosuppressant (e.g., teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No.
  • an immunosuppressant e.g., teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (A
  • the fibrosis-inhibiting compound is IMPDH (inosine monophosphate) (e.g., ribavirin (Hoffmann-La Roche) or an analogue or derivative thereof). 50. Inteqrin Antagonists
  • the fibrosis-inhibiting compound is an integrin antagonist (e.g., 683699 from Glaxo Smith Kline, integrin antagonists from Jerina AG (Germany), or an analogue or derivative thereof).
  • an integrin antagonist e.g., 683699 from Glaxo Smith Kline, integrin antagonists from Jerina AG (Germany), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an interleukin antagonist (e.g., dersalazine, or an analogue or derivative thereof).
  • an interleukin antagonist e.g., dersalazine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an interleukin 1 antagonist (e.g., NPI-1302a-3, or an analogue or derivative thereof).
  • an interleukin 1 antagonist e.g., NPI-1302a-3, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an inhibitor of type III receptor tyrosine kinase such as FLT3, PDGRF and c-KIT (e.g., MLN518 (Millenium Pharmaceuticals), or an analogue or derivative thereof).
  • type III receptor tyrosine kinase such as FLT3, PDGRF and c-KIT
  • FLT3, PDGRF and c-KIT e.g., MLN518 (Millenium Pharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an irreversible inhibitor of enzyme methionine aminopeptidase type 2 (e.g., PPI- 2458 (Praecis Pharmaceuticals), or analogue or derivative thereof).
  • methionine aminopeptidase type 2 e.g., PPI- 2458 (Praecis Pharmaceuticals), or analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an isozyme-selective delta protein kinase C inhibitor (e.g., KAI-9803 (Kai Pharmaceuticals), or an analogue or derivative thereof).
  • KAI-9803 Kai Pharmaceuticals
  • the fibrosis-inhibiting compound is a JAK3 enzyme inhibitor (e.g., CP-690,550 (Pfizer), or an analogue or derivative thereof).
  • a JAK3 enzyme inhibitor e.g., CP-690,550 (Pfizer), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a JNK inhibitor (e.g., BF-67192 (BioFocus), XG-101 or XG-102 (Xigen), or an analogue or derivative thereof).
  • JNK inhibitor e.g., BF-67192 (BioFocus), XG-101 or XG-102 (Xigen), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a kinase inhibitor (e.g., a kinase inhibitors from EVOTEC, or an analogue or derivative thereof).
  • a kinase inhibitor e.g., a kinase inhibitors from EVOTEC, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a kinesin antagonist (e.g., SB-715992 and an antifungal from Cytokinetics, or an analogue or derivative thereof).
  • a kinesin antagonist e.g., SB-715992 and an antifungal from Cytokinetics, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a leukotriene inhibitor or antagonist (e.g., ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin -beta receptor (LT- ⁇ ) from Biogen pou, Pharmaprojects No. 1535 or 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel).
  • ambicromil CAS No. 58805-38-2
  • amelubant CAS No. 346735-24-8
  • DW-1141 Denillus
  • ebselen Disaiichi Pharmaceutical
  • ibudilast ibudi
  • the fibrosis-inhibiting compound is a MAP kinase inhibitor (e.g., SRN-003-556 (Sirenade), AEG-3482 (AEgera), ARRY- 142886 (Array BioPharma), CDP-146 (UCB) or analogue or derivative thereof).
  • MAP kinase inhibitor e.g., SRN-003-556 (Sirenade), AEG-3482 (AEgera), ARRY- 142886 (Array BioPharma), CDP-146 (UCB) or analogue or derivative thereof.
  • MMPI Matrix Metalloproteinase Inhibitors
  • the fibrosis-inhibiting compound is a matrix metalloproteinase inhibitor.
  • a variety of MMPI's may be used in the practice of the invention.
  • the MMPI is a MMP-1 inhibitor.
  • the MMPI is a MMP-2 inhibitor.
  • the MMPI is a MMP-4, MMP-5, MMP-6, MMP-7, or MMP-8 inhibitor.
  • Representative examples of MMPI's include glucosamine sulfate, neovastat, GM1489 (CAS No. 170905-75-6), XL784 (EXEL-01370784), TNF-a Protease lnhibitor-1 or 2 (TAPI-1 or TAPI-2), galardin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a MCP-CCR2 inhibitor (e.g., MLN 1202 (Millennium Pharmaceuticals) , or an analogue or derivative thereof).
  • MCP-CCR2 inhibitor e.g., MLN 1202 (Millennium Pharmaceuticals) , or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an mTOR inhibitor (e.g., temsirolimus (CAS No. 162635-04-3) (Wyeth), or an analogue or derivative thereof).
  • mTOR inhibitor e.g., temsirolimus (CAS No. 162635-04-3) (Wyeth), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an mTOR kinase inhibitor (e.g., ABT-578 (Abbott), temsirolimus (Wyeth), AP- 23573 (Ariad), or an analogue or derivative thereof).
  • ABT-578 Abbott
  • temsirolimus Wyeth
  • AP- 23573 Ariad
  • the fibrosis-inhibiting compound is a microtubule inhibitor (e.g., antibody-maytansinoid conjugates from Biogen pouez, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4 or huMy9- 6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098 or IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No.
  • a microtubule inhibitor e.g., antibody-maytansinoid conjugates from Biogen pou, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-
  • the microtubule inhibitor is a microtubule polymerization inhibitor such as vincamine, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a MIF inhibitor (e.g., AVP-13546 (Avanir), an MIF inhibitor from Genzyme, migration stimulation factor D, or an analogue or derivative thereof).
  • MIF inhibitor e.g., AVP-13546 (Avanir)
  • MIF inhibitor from Genzyme e.g., Genzyme
  • migration stimulation factor D e.g., migration stimulation factor D
  • an analogue or derivative thereof e.g., a MMP (Stromolvsin) Inhibitors
  • the fibrosis-inhibiting compound is a MMP (stromolysin) inhibitor (e.g., anticancer tetracycline from Tetragenex, rhostatin (BioAxone), TIMP's from Sanofi-Aventis (CAS No. 86102-31-0), and MMP inhibitors form Cognosci and Tetragenex, or an analogue or derivative thereof).
  • MMP stromolysin
  • the fibrosis-inhibiting compound is a neurokinin (NK) antagonist (e.g., anthrotainin (CAS No. 148084-40-6) (Sanofi- Aventis), an IBS thereapeutic such as SLV-332 from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No.
  • NK neurokinin
  • SSR-240600 or SSR- 241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a NF kappa B (NFKB) inhibitor (e.g., emodin (CAS No. 518-82-1), AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
  • dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104- 49-4) (Encore Pharmaceuticals), Bay 11-7085, or an analogue or derivative thereof).
  • INDRA OXiGENE
  • IPL-576092 CAS No. 137571-30-3
  • IPL-576092 CAS No. 137571-30-3 (Inflazyme)
  • NFKB decoy Corgentech
  • NFKB decoy oligo (
  • the fibrosis-inhibiting compound is a nitric oxide agonist (e.g., Acclaim, Angx-1039 or Angx-3227 (Angiogenix), CAS-1609 (CAS No. 158590-73-9) (Sanofi-Aventis), GCI-503 (Spear Therapeutics), HCT- 3012 (CAS No. 163133-43-5) (NicOx), hydralazine + ISDN (NitroMed), isosorbide dinitrate, Diffutab (CAS No. 87-33-2) (Eurand), isosorbide mononitrate (CAS No.
  • a nitric oxide agonist e.g., Acclaim, Angx-1039 or Angx-3227 (Angiogenix), CAS-1609 (CAS No. 158590-73-9) (Sanofi-Aventis), GCI-503 (Spear Therapeutics), HCT- 3012 (CAS No. 163133-43-5) (NicOx),
  • nitroglycerin in the form of a nitroglycerin patch such as DERMATRANS from (Rottapharm), nitroglycerin (CAS No. 55-63-0) (from Cellegy Pharmaceuticals, Forest Laboratories, NovaDel, Schwarz Pharma, and Watson), NO-releasing prodrugs (Inotek), OM-294DP (OM PHARMA), oxdralazine (CAS No. 27464-23-9) (Sanofi-Aventis), pirsidomine (CAS No. 132722-74-8) (Sanofi-Aventis), prostaglandin and NO donor (Cellegy
  • upidosin derivatives (Recordati), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an ornithine decarboxylase inhibitor (e.g., aplidine, or an analogue or derivative thereof).
  • an ornithine decarboxylase inhibitor e.g., aplidine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a p38 MAP kinase inhibitor (e.g., AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74-6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), or an analogue or derivative thereof).
  • a p38 MAP kinase inhibitor e.g., AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38
  • the fibrosis-inhibiting compound is a palmitoyl-protein thioesterase inhibitor (e.g., aplidine, or an analogue or derivative thereof)-
  • a palmitoyl-protein thioesterase inhibitor e.g., aplidine, or an analogue or derivative thereof
  • the fibrosis-inhibiting compound is a PDGF receptor kinase inhibitors (e.g., AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No.
  • PDGF receptor kinase inhibitors e.g., AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No.
  • the fibrosis-inhibiting compound is a peroxisome proliferator-activated receptor (PPAR) agonists (e.g., (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, or AZD-8677 (AstraZeneca), DRF-10945 or balaglitazone (Dr Reddy's), CS-00088 or CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No.
  • PPAR peroxisome proliferator-activated receptor
  • rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), such as AVANDARYL or rosiglitazone maleate/metformin extend (CAS No. 155141-29- 0 and 657-24-9) such as AVANDAMET, or rosiglitazone maleate+metformin, such as AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), or an analogue or derivative thereof).
  • the PPAR Agonist is a PPAR ⁇ agonist such as GW7647 or fenofibric acid (CAS No. 42017-89-0), a PPAR ⁇ agonist such as MCC-555 (CAS No. 161600-01 -7), GW9662 or GW1929, a PPAER ⁇ agonist such as GW501516, a PPAR ⁇ and PPAR ⁇ agonist such L-165,041 (CAS No. 79558-09-1), or an analogue or derivative thereof.
  • a PPAR ⁇ agonist such as GW7647 or fenofibric acid (CAS No. 42017-89-0)
  • MCC-555 CAS No. 161600-01 -7
  • GW9662 or GW1929 a PPAER ⁇ agonist
  • GW501516 a PPAR ⁇ and PPAR ⁇ agonist
  • L-165,041 CAS No. 79558-09-1
  • the fibrosis-inhibiting compound is a phosphatase inhibitor (e.g., diabetes thereapy such as SQMO3, SQDM38, SQDM ⁇ O from Sequenom, Pharmaprojects No. 4191 (Sanofi-Aventis), PRL-3 inhibitors from Genzyme, WIP1 inhibitors from Amgen, or an analogue or derivative thereof).
  • a phosphatase inhibitor e.g., diabetes thereapy such as SQMO3, SQDM38, SQDM ⁇ O from Sequenom, Pharmaprojects No. 4191 (Sanofi-Aventis), PRL-3 inhibitors from Genzyme, WIP1 inhibitors from Amgen, or an analogue or derivative thereof.
  • PDE Phosphodiesterase
  • the fibrosis-inhibiting compound is a phosphodiesterase (PDE) inhibitor (e.g., avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5) or DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, or GRC-3886 (Glenmark), HWA-153 (CAS No.
  • PDE phosphodiesterase
  • the phosphodiesterase inhibitor is a phosphodiesterase III inhibitor (e.g., enoximone, or an analogue or derivative thereof).
  • the phosphodiesterase inhibitor is a phosphodiesterase IV inhibitor (e.g., fosfosal, Atopik (Barrier Therapeutics), triflusal, or an analogue or derivative thereof).
  • the phosphodiesterase inhibitor is a phosphodiesterase V inhibitor.
  • the fibrosis-inhibiting compound is a PKC inhibitor (e.g., HMR-105509 or P-10050 (Sanofi-Aventis), JNJ-10164830 (Johnson & Johnson),Ro-31-8425 (CAS No. 131848-97-0),NPC-15437 dihydrochloride (CAS No. 136449-85-9), or an analogue or derivative thereof).
  • PKC inhibitor e.g., HMR-105509 or P-10050 (Sanofi-Aventis), JNJ-10164830 (Johnson & Johnson),Ro-31-8425 (CAS No. 131848-97-0),NPC-15437 dihydrochloride (CAS No. 136449-85-9), or an analogue or derivative thereof.
  • the PKC inhibitor is an inhibitor of PKC beta (e.g., ruboxistaurin (EIi Lilly), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a platelet activating factor antagonist (e.g., dersalazine, or an analogue or derivative thereof).
  • a platelet activating factor antagonist e.g., dersalazine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a platelet-derived growth factor receptor kinase inhibitor (e.g., sorafenib tosylate, Raf or Ras inhibitors such as sorafenib tosylate from Bayer and Onyx Pharmaceuticals, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a prolyl hydroxylase inhibitor (e.g., FG-2216 (CAS No. 11096-26-7) or HIF agonists from FibroGen, or an analogue or derivative thereof).
  • FG-2216 CAS No. 11096-26-7
  • HIF agonists from FibroGen or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a polymorphonuclear neutrophil inhibitor (e.g., orazipone, or an analogue or derivative thereof).
  • a polymorphonuclear neutrophil inhibitor e.g., orazipone, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a protein kinase B inhibitor (e.g., Akt-1 inhibitors from Amphora, or an analogue or derivative thereof).
  • Akt-1 inhibitors from Amphora, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a protein kinase C stimulant (e.g., bryostatin-1 , or analogue or derivative thereof).
  • a protein kinase C stimulant e.g., bryostatin-1 , or analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a purine nucleoside analogue (e.g., cladrinbine and formulations thereof, such as MYLINAX from Serone SA and IVAX Research Inc. (Miami, FL), or an analogue or derivative thereof).
  • a purine nucleoside analogue e.g., cladrinbine and formulations thereof, such as MYLINAX from Serone SA and IVAX Research Inc. (Miami, FL), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a purinoreceptor P2X antagonist (e.g., AZD-9056 (AstraZeneca), R-1554 (Hoffmann-La Roche), AR-C118925XX (AstraZeneca), suramin (CAS No. 129- 46-4), P2Y4 receptor from Euroscreen, or an analogue or derivative thereof).
  • AZD-9056 AstraZeneca
  • R-1554 Hoffmann-La Roche
  • AR-C118925XX AstraZeneca
  • suramin CAS No. 129- 46-4
  • P2Y4 receptor from Euroscreen, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a Raf kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
  • a Raf kinase inhibitor e.g., sorafenib tosylate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a reversible inhibitor (e.g., lapatinib (GSK), or an analogue or derivative thereof).
  • GSK lapatinib
  • the fibrosis-inhibiting compound is a cytoplasmic tyrosine kinase inhibitor such as a SRC inhibitor (e.g., SRN-004 (Sirenade), gallium maltolate (Titan Pharmaceutcals), or an analogue or derivative thereof), or an analogue or derivative thereof).
  • SRC inhibitor e.g., SRN-004 (Sirenade), gallium maltolate (Titan Pharmaceutcals), or an analogue or derivative thereof), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a SDF-1 antagonist (e.g., CTCE-9908 (Chemokine Therapeutics), or an analogue or derivative thereof).
  • SDF-1 antagonist e.g., CTCE-9908 (Chemokine Therapeutics), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a sheddase inhibitor (e.g., INCB-7839 (Incyte Corporation), or an analogue or derivative thereof).
  • a sheddase inhibitor e.g., INCB-7839 (Incyte Corporation)
  • an analogue or derivative thereof e.g., INCB-7839 (Incyte Corporation)
  • the fibrosis-inhibiting compound is a SRC inhibitor (e.g., SRN-004 (Sirenade), or an analogue or derivative thereof).
  • the SRC inhibitor is a SRC kinase inhibitor (e.g., AZD0530 (AstraZeneca), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a stromelysin inhibitor (e.g., glucosamine sulfate, or an analogue or derivative thereof).
  • a stromelysin inhibitor e.g., glucosamine sulfate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a syk kinase inhibitor (e.g., R406 (Rigel), or an analogue or derivative thereof).
  • a syk kinase inhibitor e.g., R406 (Rigel), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a telomerase inhibitor (e.g., AS-1410 (Antisoma), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a TGF beta inhibitor (e.g., pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF- ⁇ antagonists from Inflazyme (Pharmaprojects No. 6075), TGF- ⁇ antagonists (e.g., 1090 and 1091 from Sydney; non-industrial source), TGF- ⁇ I receptor kinase inhibitors from EIi Lilly, TGF- ⁇ receptor inhibitors from Johnson & Johnson, or an analogue or derivative thereof).
  • TNF ⁇ Antagonists and TACE Inhibitors e.g., pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-
  • the fibrosis-inhibiting compound is a TNF ⁇ antagonist or TACE inhibitors (e.g., adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No.
  • TNF ⁇ antagonist or TACE inhibitors e.g., adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha ki
  • TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
  • TNF antagonist from Jerina AG (Germany), dersalazine, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a tumor necrosis factor (TNF) antagonist (e.g., anti-inflammatory compounds from Biota Inc., or an analogue or derivative thereof).
  • TNF tumor necrosis factor
  • the fibrosis-inhibiting compound is a Toll receptor antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof).
  • a Toll receptor antagonist e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a tubulin antagonist (e.g., synthadotin, KRX-0403 (Keryx Biopharmaceuticals), or an analogue or derivative thereof).
  • a tubulin antagonist e.g., synthadotin, KRX-0403 (Keryx Biopharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a tyrosine kinase inhibitor (e.g., SU-011248 (e.g., SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), (e.g., AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim),
  • EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax- EGFr (Genmab), idronoxil (CAS No.
  • the tyrosine kinase inhibitor is an EGFR tyrosine kinase inhibitor such as EKB-569 (Wyeth), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a VEGF Inhibitor (e.g., AZD2171 (AstraZeneca), or an analogue or derivative thereof).
  • a VEGF Inhibitor e.g., AZD2171 (AstraZeneca), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a vitamin D receptor agonist (e.g., BXL-628, BXL-922 (BioXell), or an analogue or derivative thereof).
  • a vitamin D receptor agonist e.g., BXL-628, BXL-922 (BioXell), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an histamine receptor antagonist.
  • the histamine receptor antagonists such as H1 , H2, and H3 histamine receptor antagonists, block the production of pro-inflammatory cytokines such as TNFa and IL-1 (e.g., IL- 1 ⁇ ).
  • the histamine receptor antagonist inhibit NFkB activation.
  • H1 histamine receptor antagonists include phenothiazines, such as promethazine, and alkylamines, such as chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No.
  • histamine receptor antagonists include broad spectrum histamine receptor antagonists such as methylxanthines (e.g., theophylline, theobromine, and caffeine).
  • H2 receptor antagonists include those with a histamine-like structure including cimetidine (available under the tradename TAGAMET from SmithKline Beecham Pbamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ) 1 famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1).
  • cimetidine available under the tradename TAGAMET from SmithKline Beecham Pbamaceutical Co., Wilmington, DE
  • ranitidine available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ
  • 1 famotidine available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ
  • nizatidine available under the tradename AXID
  • H3 receptor antagonists e.g., thioperamide and thioperamide maleate salt
  • antihistamines such as tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones.
  • the fibrosis-inhibiting compound is an alpha adrenergic receptor antagonist.
  • Alpha adrenergic receptor antagonists may inhibit the production of pro-inflammatory cytokines such as TNFa.
  • the alpha adrenergic receptor antagonist may be an alpha-1 and/or an alpha-2 adrenergic receptor antagonist.
  • Representative examples of alpha-1/alpha-2 antagonists include phenoxybenzamine.
  • the alpha adrenergic receptor antagonist is a haloalkylamine compound or a catecholamine uptake inhibitor.
  • Representative examples of alpha-1 adrenergic receptor antagonists include phenoxybenzamine hydrochloride and prazosin, a piperizinyl quinazoline.
  • alpha-2 adrenergic receptor antagonists include imadazole based compounds such as idazoxan (CAS No. 79944-56-2), idazoxan hydrochloride, and loxapine succinate salt (CAS No. 27833-64-3). Additional examples of alpha adrenergic receptor antagonists include prazosin hydrochloride.
  • the fibrosis-inhibiting compound is an anti-psychotic compound, such as a phenothiazine compound or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a phenothiazine derivative capable of suppressing the production of proinflammatory cytokines such as TNFa and/or IL-I .
  • phenothiazine compounds include chlorpromazine, fluphenazine, trifluorphenazine, mesoridazine, thioridazine, and perphenazine.
  • anti-psychotic compounds include thioxanthines such as chlorprothixene and thiothixene, clozapine, loxapine succinate, and olanzapine.
  • the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is G protein agonist, such as aluminum fluoride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an antibiotic, such as apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, or an analogue or derivative thereof.
  • antibiotic such as apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-microbial agent, such as brefeldin A (CAS No. 20350-15-6), terbinafine, benzoyl peroxide, pentamidine, ornidazole, tinidazole, ketocanazole, sulconazole nitrate salt, or an analogue or derivative thereof.
  • brefeldin A CAS No. 20350-15-6
  • terbinafine benzoyl peroxide
  • pentamidine ornidazole
  • tinidazole ketocanazole
  • sulconazole nitrate salt or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is DNA topoisomerase I inhibitor, such as ⁇ -lapachone (CAS No. 4707-32-8), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is DNA topoisomerase Il inhibitor, such as (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is thromboxane A2 receptor inhibitor, such as BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), or an analogue or derivative thereof.
  • thromboxane A2 receptor inhibitor such as BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a D2 dopamine receptor antagonist, such as clozapine (CAS No. 5786-21-0), mesoridazine benzenesulfonate, or an analogue or derivative thereof.
  • D2 dopamine receptor antagonist such as clozapine (CAS No. 5786-21-0), mesoridazine benzenesulfonate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, such as juglone (CAS No. 481- 39-0), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a dopamine antagonist, such as thiothixene, thioridazine hydrochloride, or an analogue or derivative thereof. 116) Anesthetics
  • the fibrosis-inhibiting compound is an anesthetic compound, such as lidocaine (CAS No. 137-58-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a clotting factor, such as menadione (CAS No. 58-27-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a lysyl hydrolase inhibitor, such as minoxidil (CAS No. 38304-91-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a muscarinic receptor inhibitor, such as perphenazine (CAS No. 58-39-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a superoxide anion generator, such as plumbagin (CAS No. 481-42-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a steroid, such as prednisolone 21-acetate (CAS No. 52-21-1), loteprednol etabonate, (CAS No. 82034-46-6), clobetasol propionate, or an analogue or derivative thereof. 122”) Anti-Proliferative Agents
  • the fibrosis-inhibiting compound is an anti-proliferative agent, such as silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2-hexanediol, dioctyl phthalate (CAS No.
  • zirconium (IV) oxide zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride or tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methy!-L- arginine acetate salt, galardin, halofuginone hydrobromide (HBr), fascaplysin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a diuretic, such as spironolactone (CAS No. 52-01-7), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-coagulant, such as fucoidan from Fucus vesiculosus (CAS No. 9072-19-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cyclic GMP agonist, such as sinitrodil (CAS No. 143248-63-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an adenylate cyclase agonist, such as histamine (CAS No. 51-45-6), or an analogue or derivative thereof. 127) Antioxidants
  • the fibrosis-inhibiting compound is an antioxidant, such as morpholine, phytic acid dipotassium salt, (-)- epigallocatechin or (-)-epigallocatechin gallate from green tea (CAS Nos. 970- 74-1 and 1257-08-5, respectively), (-)-epigallocatechin gallate (CAS No. 989- 51-5), nobiletin (CAS No. 478-01-3), probucol (CAS No. 23288-49-5), phosphorous acid, hesperetin, L-ascorbyl-2-phosphate, magnesium salt (CAS No. 84309-23-9), catechin, ( ⁇ )-naringenin (CAS No. 67604-48-2), (-)- epicatechin, 3-hydroxyflavone, (-)-epicatechin gallate, (-)-arctigenin, or an analogue or derivative thereof.
  • an antioxidant such as morpholine, phytic acid dipotassium salt, (-)
  • the fibrosis-inhibiting compound is a nitric oxide synthase inhibitor, such as ammonium pyrrolidinedithiocarbamate (CAS No. 5108-96-3), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a reversible nitric oxide synthase inhibitor, such as NB-methyl-L-arginine acetate salt (L-NMMA) (CAS No. 53308-83-1), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an antineoplastic agent, such as tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine (CAS Number: 4291-63-8; Leustat®, Janssen-Ortho), imatinib mesilate, or an analogue or derivative thereof.
  • antineoplastic agent such as tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine (CAS Number: 4291-63-8; Leustat®, Janssen-Ortho), imatinib mesilate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a DNA synthesis inhibitor, such as S-(2-hydroxy-5-nitrobenyl)-6-thioguanosine or uracilfludarabine phosphate (CAS No. 75607-67-9), 6,11-dihydroxy-5,12- naphthacenedione, cladribine (CAS Number: 4291-63-8; Leustat®, Janssen- Ortho), or an analogue or derivative thereof. 131) DNA Alkylating A ⁇ ents
  • the fibrosis-inhibiting compound is a DNA alkylating agent, such as dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, or an analogue or derivative thereof.
  • a DNA alkylating agent such as dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a DNA methylation inhibitor, such as decitabine (CAS No. 2353-33-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a
  • NSAID agent such as nabumetone, benzydamine hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, such as trans- styrylacetic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a MEK1/MEK 2 inhibitor, such as U0126 (CAS No. 109511-58-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an NO synthase inhibitor, such as L-NAME (CAS No. 53308-83-1), NG-Methyl-L- arginine acetate salt, or an analogue or derivative thereof.
  • NO synthase inhibitor such as L-NAME (CAS No. 53308-83-1), NG-Methyl-L- arginine acetate salt, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is retinoic acid receptor antagonist, such as isotretinoin (CAS No. 4759-48-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an ACE inhibitor, such as quinapril hydrochloride (CAS No. 85441-61-8), enalapril, or an analogue or derivative thereof.
  • ACE inhibitor such as quinapril hydrochloride (CAS No. 85441-61-8), enalapril, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a glycosylation inhibitor, such as aminoguanidine hydrochloride, castanospermine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an intracellular calcium influx inhibitor, such as TAS-301 (CAS No. 193620-69-8), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-emetic agent, such as amifostine (CAS No. 20537-88-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an acetylcholinesterase inhibitor, such as (-)-huperzine A (CAS No. 102518-79-6), or an analogue or derivative thereof. 143) ALK-5 Receptor Antagonists
  • the fibrosis-inhibiting compound is an ALK-5 receptor antagonist, such as SB 431542 (CAS No. 301836-41-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a RAR/RXT antagonist, such as 9-cis-retinoic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a elF-
  • the fibrosis-inhibiting compound is a S- adenosyl-L-homocysteine hydrolase inhibitor, such as 3-deazaadenosine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an estrogen agonist, such as coumestrol, bisphenol A, 1-Iinoleoyl-rac-glycerol (CAS No. 2277-28-3), daidzein (4,7-dihydroxy-iso-flavone), dihexyl phthalate, kaempferol, formononetin, , or an analogue or derivative thereof.
  • estrogen agonist such as coumestrol, bisphenol A, 1-Iinoleoyl-rac-glycerol (CAS No. 2277-28-3), daidzein (4,7-dihydroxy-iso-flavone), dihexyl phthalate, kaempferol, formononetin, , or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a serotonin receptor inhibitor, such as amitriptyline hydrochloride, or an analogue or derivative thereof. 149) Anti-Thrombotic Agents
  • the fibrosis-inhibiting compound is an anti-thrombotic agent, such as geniposidic acid, geniposide, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a tryptase inhibitors, such as 2-azetidinone, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a pesticide, such as allyl disulfide, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a bone mineralization promotor, such as glycerol 2-phosphate disodium salt hydrate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a bisphosphonate compound, such as risedronate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-inflammatory compound, such as aucubin, cepharanthine, or an analogue or derivative thereof. 155) DNA Methylation Promotors
  • the fibrosis-inhibiting compound is a DNA methylation promotor, such as 5-azacytidine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-spasmodic agent, such as 2-hydroxy-4,6-dimethoxyacetophenone, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a protein synthesis inhibitor, such as oxytetracycline hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a ⁇ - glucosidase inhibitor, such as myricetin (CAS No. 529-44-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a calcium channel blocker, such as verapamil, nitrendipine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a L- type calcium channel blocker, such as nifedipine (CAS No. 21829-25-4), (+)-cis- diltiazem hydrochloride, or an analogue or derivative thereof.
  • L- type calcium channel blocker such as nifedipine (CAS No. 21829-25-4), (+)-cis- diltiazem hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a T- type calcium channel blocker, such as penfluridol (CAS No. 26864-56-2), or an analogue or derivative thereof. 160) Pyruvate Dehydrogenase Activators
  • the fibrosis-inhibiting compound is a pyruvate dehydrogenase activator, such as dichloroacetic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a prostaglandin inhibitor, such as betulinic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a sodium channel inhibitor, such as amiloride hydrochloride hydrate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a serine protease inhibitor, such as gabexate mesylate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an intracellular calcium flux inhibitor, such as thapsigargin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a JAK2 inhibitor (e.g., AG-490 (CAS No. 134036-52-5), or an analogue or derivative thereof). 166) Androgen Inhibitors
  • the fibrosis-inhibiting compound is an androgen inhibitor (e.g., tibolone (CAS No. 5630-53-5), or an analogue or derivative thereof).
  • an androgen inhibitor e.g., tibolone (CAS No. 5630-53-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an aromatase inhibitor (e.g., letrozole, or an analogue or derivative thereof).
  • an aromatase inhibitor e.g., letrozole, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-viral agent, such as imiquimod, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a 5-HT inhibitor, such as ketanserin tartrate, amoxapine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a FXR antagonist, such as guggulsterone (CAS No. 95975-55-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an actin polymerization and stabilization promotor, such as jasplakinolide, or an analogue or derivative thereof. 172) AXOR12 Agonists
  • the fibrosis-inhibiting compound is an AXOR12 agonist, such as metastin (KiSS-1 (112-121), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an angiotensin Il receptor agonist, such as losartan potassium, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a platelet aggregation inhibitor, such as clopidogrel, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a CB1/CB2 receptor agonist, such as HU-210 (CAS No. 112830-95-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a norepinephrine reuptake inhibitor, such as nortriptyline hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a selective serotonin reuptake inhibitor, such as paroxetine maleate, or an analogue or derivative thereof. 178) Reducing Agents
  • the fibrosis-inhibiting compound is a reducing agent such as WW-85 (lnotek), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an immunomodulators such as Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, or an analogue or derivative thereof.Combination Therapies
  • compositions may further include a compound which acts to have an inhibitory effect on pathological processes in or around the treatment site.
  • additional therapeutically active agents include, by way of example and not limitation, anti-thrombotic agents, anti-proliferative agents, antiinflammatory agents, neoplastic agents, enzymes, receptor antagonists or agonists, hormones, antibiotics, antimicrobial agents, antibodies, cytokine inhibitors, IMPDH (inosine monophosplate dehydrogenase) inhibitors tyrosine kinase inhibitors, MMP inhibitors, p38 MAP kinase inhibitors, immunosuppressants, apoptosis antagonists, caspase inhibitors, and JNK inhibitors.
  • anti-thrombotic agents include, by way of example and not limitation, anti-thrombotic agents, anti-proliferative agents, antiinflammatory agents, neoplastic agents, enzymes, receptor antagonists or agonists, hormones, antibiotics, antimicrobial agents, antibodies, cytokine inhibitors, IMPDH (inosine monophosplate dehydrogenase) inhibitors tyrosine kinas
  • the present invention also provides for the combination of an implantable pump or implantable sensor device (as well as compositions and methods for making implantable pump and sensor devices) that includes an anti-fibrosing agent and an anti-infective agent, which reduces the likelihood of infections.
  • Infection is a common complication of the implantation of foreign bodies such as, for example, medical devices.
  • Foreign materials provide an ideal site for micro- organisms to attach and colonize. It is also hypothesized that there is an impairment of host defenses to infection in the microenvironment surrounding a foreign material. These factors make medical implants particularly susceptible to infection and make eradication of such an infection difficult, if not impossible, in most cases.
  • the present invention provides agents (e.g., chemotherapeutic agents) that can be released from a composition, and which have potent antimicrobial activity at extremely low doses.
  • a wide variety of anti-infective agents can be utilized in combination with the present compositions. Suitable anti-infective agents may be readily determined based the assays provided in Example 51.
  • agents that can be used: (A) anthracyclines ⁇ e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin).
  • fluoropyrimidines e.g., 5-FU
  • C folic acid antagonists
  • D podophylotoxins
  • E camptothecins
  • F hydroxyureas
  • platinum complexes e.g., cisplatin
  • Anthracyclines have the following general structure, where the R groups may be a variety of organic groups:
  • R 1 is CH 3 or CH 2 OH
  • R 2 is daunosamine or H
  • R 3 and R 4 are independently one of OH, NO 2 , NH 2 , F, Cl, Br, I, CN, H or groups derived from these
  • R5 is hydrogen, hydroxyl, or methoxy
  • R 6-S are all hydrogen.
  • R 5 and R 6 are hydrogen and R 7 and R 8 are alkyl or halogen, or vice versa.
  • R 1 may be a conjugated peptide.
  • R 5 may be an ether linked alkyl group.
  • R 5 may be OH or an ether linked alkyl group.
  • Ri may also be linked to the anthracycline ring by a group other than C(O), such as an alkyl or branched alkyl group having the C(O) linking moiety at its end, such as -CH 2 CH(CH 2 -X)C(O)-R 1 , wherein X is H or an alkyl group (see, e.g., U.S. Patent 4,215,062).
  • R 3 may have the following structure:
  • Ri 0 may be H or form a secondary amine with a group such as an aromatic group, saturated or partially saturated 5 or 6 membered heterocyclic having at least one ring nitrogen (see U.S. Patent 5,843,903).
  • Ri 0 may be derived from an amino acid, having the structure -
  • Ri 2 may be H, alkyl, aminoalkyl, amino, hydroxyl, mercapto, phenyl, benzyl or methylthio (see U.S. Patent 4,296,105).
  • anthracyclines are doxorubicin, daunorubicin, idambicin, epirubicin, pirarubicin, zorubicin, and carubicin.
  • Suitable compounds have the structures:
  • Doxorubicin OCH 3 C(O)CH 2 OH OH out of ring plane
  • Epirubicin (4' epimer of OCH 3 C(O)CH 2 OH OH in ring plane doxorubicin)
  • Daunorubicin OCH 3 C(O)CH 3 OH out of ring plane
  • Idarubicin H C(O)CH 3 OH out of ring plane
  • Pirarubicin OCH 3 C(O)CH 2 OH ⁇ - 1
  • Carubicin OH C(O)CH 3 OH out of ring plane
  • anthracyclines are anthramycin, mitoxantrone, menogaril, nogalamycin, aclacinomycin A, olivomycin A, chromomycin A 3 , and plicamycin having the structures:
  • Plicamycin H H H CH 3 Other representative anthracyclines include, FCE 23762, a doxorubicin derivative (Quaglia et al., J. Liq. Chromatogr. 17(18):3911-3923, 1994), annamycin (Zou et al., J. Pharm. ScL 82(11):1151-1154, 1993), ruboxyl (Rapoport ef al., J. Controlled Release 58(2): 153- 162, 1999), anthracycline disaccharide doxorubicin analogue (Pratesi et al., Clin. Cancer Res.
  • deoxydihydroiodooxorubicin EPA 275966
  • adriblastin Kalishevskaya et al., Vestn. Mosk. Univ., 16(B ⁇ o ⁇ . 1):21-7, 1988
  • 4'-deoxydoxorubicin Schoelzel et al., Leuk. Res. 70(12): 1455-9, 1986
  • 4-demethyoxy-4'-o-methyldoxorubicin (Giuliani et ai, Proc. Int. Congr. Chemother.
  • the therapeutic agent is a fluoropyrimidine analog, such as 5-fluorouracil, or an analogue or derivative thereof, including carmofur, doxifluridine, emitefur, tegafur, and floxuridine.
  • fluoropyrimidine analog such as 5-fluorouracil
  • an analogue or derivative thereof including carmofur, doxifluridine, emitefur, tegafur, and floxuridine.
  • Exemplary compounds have the structures:
  • fluoropyrimidine analogues include 5-FudR (5- fluoro-deoxyuridine), or an analogue or derivative thereof, including 5- iododeoxyuridine (5-ludR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP).
  • 5-FudR 5- fluoro-deoxyuridine
  • an analogue or derivative thereof including 5- iododeoxyuridine (5-ludR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP).
  • Exemplary compounds have the structures:
  • fluoropyrimidine analogues include N3-alkylated analogues of 5-fluorouracil (Kozai et al., J. Chem. Soc, Perkin Trans. 7(19):3145-3146, 1998), 5-fluorouracil derivatives with 1 ,4- oxaheteroepane moieties (Gomez et al., Tetrahedron 54(43):13295-13312, 1998), 5-fluorouracil and nucleoside analogues (Li, Anticancer Res.
  • the therapeutic agent is a folic acid antagonist, such as methotrexate or derivatives or analogues thereof, including edatrexate, trimetrexate, raltitrexed, piritrexim, denopterin, tomudex, and pteropterin.
  • Methotrexate analogues have the following general structure:
  • R group may be selected from organic groups, particularly those groups set forth in U.S. Patent Nos. 5,166,149 and 5,382,582.
  • Ri may be N
  • R 2 may be N or C(CH 3 )
  • R 3 and R 3 ' may H or alkyl, e.g., CH 3
  • R 4 may be a single bond or NR, where R is H or alkyl group.
  • 6 , 8 may be H, OCH 3 , or alternately they can be halogens or hydro groups.
  • R 7 is a side chain of the general structure:
  • the carboxyl groups in the side chain may be esterified or form a salt such as a Zn 2+ salt.
  • Rg and R 10 can be NH 2 or may be alkyl substituted.
  • Exemplary folic acid antagonist compounds have the structures:
  • N-( ⁇ -aminoacyl) methotrexate derivatives Cheung et al., Pteridines 3( 1-2): 101 -2, 1992
  • biotin methotrexate derivatives Fean et al. , Pteridines 3(1 -2): 131 -2, 1992
  • D-glutamic acid or D-erythrou threo-4-fluoroglutamic acid methotrexate analogues
  • the therapeutic agent is a podophyllotoxin, or a derivative or an analogue thereof.
  • exemplary compounds of this type are etoposide or teniposide, which have the following structures:
  • podophyllotoxins include Cu(II)- VP-16 (etoposide) complex (Tawa et al., Bioorg. Med. Chem. 6(7):1003-1008, 1998), pyrrolecarboxamidino-bearing etoposide analogues (Ji et al., Bioorg. Med. Chem. Lett. 7(5):607-612, 1997), 4 ⁇ -amino etoposide analogues (Hu, University of North Carolina Dissertation, 1992), ⁇ -lactone ring-modified arylamino etoposide analogues (Zhou et al., J. Med. Chem.
  • the therapeutic agent is camptothecin, or an analogue or derivative thereof.
  • Camptothecins have the following general structure.
  • X is typically O, but can be other groups, e.g., NH in the case of 21-Iactam derivatives.
  • Ri is typically H or OH, but may be other groups, e.g., a terminally hydroxylated Ci_ 3 alkane.
  • R2 is typically H or an amino containing group such as (CH 3 ) 2 NHCH 2 , but may be other groups e.g., NO 2 , NH 2 , halogen (as disclosed in, e.g., U.S. Patent 5,552,156) or a short alkane containing these groups.
  • R 3 is typically H or a short alkyl such as C 2 H 5 .
  • R 4 is typically H but may be other groups, e.g., a methylenedioxy group with Ri .
  • camptothecin compounds include topotecan, irinotecan (CPT-11), 9-aminocamptothecin, 21-lactam-20(S)-camptothecin, lO.H-methylenedioxycamptothecin, SN-38, 9-nitrocamptothecin, 10- hydroxycamptothecin.
  • Exemplary compounds have the structures:
  • Camptothecins have the five rings shown here.
  • the ring labeled E must be intact (the lactone rather than carboxylate form) for maximum activity and minimum toxicity.
  • Camptothecins are believed to function as topoisomerase I inhibitors and/or DNA cleavage agents.
  • the therapeutic agent of the present invention may be a hydroxyurea.
  • Hydroxyureas have the following general structure:
  • Suitable hydroxyureas are disclosed in, for example, U.S. Patent No. 6,080,874, wherein R 1 is:
  • R 2 is an alkyl group having 1-4 carbons and R 3 is one of H, acyl, methyl, ethyl, and mixtures thereof, such as a methylether.
  • Ri is a cycloalkenyl group, for example N-[3-[5-(4- fluorophenylthio)-furyl]-2-cyclopenten-1-yl]N-hydroxyurea; R 2 is H or an alkyl group having 1 to 4 carbons and R 3 is H; X is H or a cation.
  • Suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 4,299,778, wherein Ri is a phenyl group substituted with one or more fluorine atoms; R 2 is a cyclopropyl group; and R 3 and X is H.
  • n is 0-2 and Y is an alkyl group.
  • the hydroxyurea has the structure:
  • the therapeutic agent is a platinum compound.
  • suitable platinum complexes may be of Pt(II) or Pt(IV) and have this basic structure:
  • X and Y are anionic leaving groups such as sulfate, phosphate, carboxylate, and halogen; R 1 and R 2 are alkyl, amine, amino alkyl any may be further substituted, and are basically inert or bridging groups.
  • Z 1 and Z 2 are non-existent.
  • Z 1 and Z 2 may be anionic groups such as halogen, hydroxy, carboxylate, ester, sulfate or phosphate. See, e.g., U.S. Patent Nos. 4,588,831 and 4,250,189.
  • Suitable platinum complexes may contain multiple Pt atoms. See, e.g., U.S. Patent Nos. 5,409,915 and 5,380,897.
  • platinum compounds are cisplatin, carboplatin, oxaliplatin, and miboplatin having the structures:
  • platinum compounds include (CPA) 2 Pt[DOLYM] and (DACH)Pt[DOLYM] cisplatin (Choi et al., Arch. Pharmacal Res. 22(2): 151 -156, 1999), Cis-[PtCI 2 (4,7-H-5-methyl-7- oxo]1 ,2,4[triazolo[1,5-a]pyrimidine) 2 ] (Navarro et al., J. Med. Chem. 41 (3):332- 338, 1998), [Pt(cis-1 ,4-DACH)(trans-CI 2 )(CBDCA)] • 1 / 2 MeOH cisplatin (Shamsuddin et al., Inorg. Chem.
  • the total dose of doxorubicin applied to the implant should not exceed 25 mg (range of 0.1 ⁇ g to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 ⁇ g to 5 mg.
  • the dose per unit area (Ae., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 ⁇ g - 100 ⁇ g per mm 2 of surface area.
  • doxorubicin should be applied to the implant surface at a dose of 0.1 ⁇ g/mm 2 - 10 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10 "8 - 10 "4 M of doxorubicin is maintained on the surface. It is necessary to insure that surface drug concentrations exceed concentrations of doxorubicin known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10 "4 M; although for some embodiments lower concentrations are sufficient).
  • doxorubicin is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of doxorubicin (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as doxorubicin is administered at half the above parameters, a compound half as potent as doxorubicin is administered at twice the above parameters, etc.).
  • the total dose of mitoxantrone applied should not exceed 5 mg (range of 0.01 ⁇ g to 5 mg).
  • the total amount of drug applied should be in the range of 0.1 ⁇ g to 3 mg.
  • the dose per unit area i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated
  • mitoxantrone should be applied to the implant surface at a dose of 0.05 ⁇ g/mm 2 - 5 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10 "4 - 10 "8 M of mitoxantrone is maintained. It is necessary to insure that drug concentrations on the implant surface exceed concentrations of mitoxantrone known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10 "5 M; although for some embodiments lower drug levels will be sufficient).
  • mitoxantrone is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of mitoxantrone (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as mitoxantrone is administered at half the above parameters, a compound half as potent as mitoxantrone is administered at twice the above parameters, etc.).
  • the total dose of 5-fluorouracil applied should not exceed 250 mg (range of 1.0 ⁇ g to 250 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 10 ⁇ g to 25 mg.
  • the dose per unit area i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated
  • 5-fluorouracil should be applied to the implant surface at a dose of 0.5 ⁇ g/mm 2 - 50 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10 "4 - 10 "7 M of 5-fluorouracil is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of 5-fluorouracil known to be lethal to numerous species of bacteria and fungi (i.e., are in excess of 10 "4 M; although for some embodiments lower drug levels will be sufficient).
  • 5-fluorouracil is released from the implant surface such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of 5- fluorouracil (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as 5- fluorouracil is administered at half the above parameters, a compound half as potent as 5-fluorouracil is administered at twice the above parameters, etc.).
  • Podophylotoxins Podophylotoxins.
  • the total dose of etoposide applied should not exceed 25 mg (range of 0.1 ⁇ g to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 ⁇ g to 5 mg.
  • the dose per unit area i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated
  • etoposide should be applied to the implant surface at a dose of 0.1 ⁇ g/mm 2 - 10 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a concentration of 10 "4 - 10 '7 M of etoposide is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of etoposide known to be lethal to a variety of bacteria and fungi (i.e., are in excess of 10 "s M; although for some embodiments lower drug levels will be sufficient).
  • etoposide is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of etoposide (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as etoposide is administered at half the above parameters, a compound half as potent as etoposide is administered at twice the above parameters, etc.).
  • anthracyclines e.g., doxorubicin or mitoxantrone
  • fluoropyrimidines e.g., 5-fluorouracil
  • folic acid antagonists e.g., methotrexate and/or podophylotoxins (e.g., etoposide)
  • podophylotoxins e.g., etoposide
  • an anti-infective agent e.g., anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate and/or podophylotoxins (e.g., etoposide)
  • anthracyclines e.g., doxorubicin or mitoxantrone
  • fluoropyrimidines e.g., 5-fluorouracil
  • folic acid antagonists e.g., methotrexate and/or podophylotoxins (e.g., etoposide)
  • traditional antibiotic and/or antifungal agents e.g., doxorubicin or mitoxantrone
  • fluoropyrimidines e.g., 5-fluorouracil
  • folic acid antagonists e.g., methotrex
  • the anti-infective agent may be further combined with anti-thrombotic and/or antiplatelet agents (for example, heparin, dextran sulphate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, aspirin, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, ticlopidine, clopidogrel, abcixamab, eptifibatide, tirofiban, streptokinase, and/or tissue plasminogen activator) to enhance efficacy.
  • anti-thrombotic and/or antiplatelet agents for example, heparin, dextran sulphate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, aspirin, phenylbutazone
  • one or more other pharmaceutically active agents can be incorporated into the present compositions and devices to improve or enhance efficacy.
  • additional therapeutically active agents include, by way of example and not limitation, anti-thrombotic agents, anti-proliferative agents, antiinflammatory agents, neoplastic agents, enzymes, receptor antagonists or agonists, hormones, antibiotics, antimicrobial agents, antibodies, cytokine inhibitors, IMPDH (inosine monophosplate dehydrogenase) inhibitors tyrosine kinase inhibitors, MMP inhibitors, p38 MAP kinase inhibitors, immunosuppressants, apoptosis antagonists, caspase inhibitors, and JNK inhibitors.
  • anti-thrombotic agents include, by way of example and not limitation, anti-thrombotic agents, anti-proliferative agents, antiinflammatory agents, neoplastic agents, enzymes, receptor antagonists or agonists, hormones, antibiotics, antimicrobial agents, antibodies, cytokine inhibitors, IMPDH (inosine monophosplate dehydrogenase) inhibitors tyrosine kinas
  • Implantable implantable pump and sensor devices and compositions for use with implantable pump and sensor devices may further include an anti-thrombotic agent and/or antiplatelet agent and/or a thrombolytic agent, which reduces the likelihood of thrombotic events upon implantation of a medical implant.
  • a device is coated on one aspect with a composition which inhibits fibrosis (and/or restenosis), as well as being coated with a composition or compound which prevents thrombosis on another aspect of the device.
  • anti-thrombotic and/or antiplatelet and/or thrombolytic agents include heparin, heparin fragments, organic salts of heparin, heparin complexes (e.g., benzalkonium heparinate, tridodecylammonium heparinate), dextran, sulfonated carbohydrates such as dextran sulphate, Coumadin, coumarin, heparinoid, danaparoid, argatroban chitosan sulfate, chondroitin sulfate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, acetylsalicylic acid, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, streptokinase, factor Xa inhibitors, such as DXa inhibitor
  • Further examples include plasminogen, lys-plasminogen, alpha-2-antiplasmin, urokinase, aminocaproic acid, ticlopidine, clopidogrel, trapidil (triazolopyrimidine), naftidrofuryl, auriritricarboxylic acid and glycoprotein llb/llla inhibitors such as abcixamab, eptifibatide, and tirogiban.
  • agents capable of affecting the rate of clotting include glycosaminoglycans, danaparoid, 4- hydroxycourmarin, warfarin sodium, dicumarol, phenprocoumon, indan-1 ,3- dione, acenocoumarol, anisindione, and rodenticides including bromadiolone, brodifacoum, diphenadione, chlorophacinone, and pidnone.
  • compositions for use with implantable pump and sensor devices may be or include a hydrophilic polymer gel that itself has anti-thrombogenic properties.
  • the composition can be in the form of a coating that can comprise a hydrophilic, biodegradable polymer that is physically removed from the surface of the device over time, thus reducing adhesion of platelets to the device surface.
  • the gel composition can include a polymer or a blend of polymers.
  • the anti-thrombotic composition can include a crosslinked gel formed from a combination of molecules (e.g., PEG) having two or more terminal electrophilic groups and two or more nucleophilic groups.
  • Implantable pump and sensor devices and compositions for use with implantable pump and sensor devices may further include a compound which acts to have an inhibitory effect on pathological processes in or around the treatment site.
  • the agent may be selected from one of the following classes of compounds: anti-inflammatory agents (e.g., dexamethasone, cortisone, fludrocortisone, prednisone, prednisolone, 6 ⁇ - methylprednisolone, triamcinolone, betamethasone, and aspirin); MMP inhibitors (e.g., batimistat, marimistat, TIMP's representative examples of which are included in U.S. Patent Nos. 5,665,777; 5,985,911 ; 6,288,261 ;
  • anti-inflammatory agents e.g., dexamethasone, cortisone, fludrocortisone, prednisone, prednisolone, 6 ⁇ - methylprednisolone, triamcinolone, betamethasone
  • IMPDH inosine monophosplate dehydrogenase inhibitors
  • mycophenolic acid ribaviran, aminothiadiazole, thiophenfurin, tiazofurin, viramidine
  • WO 00/63204A2 WO 01/21591 A1 , WO 01/35959A1 , WO 01/74811A2, WO 02/18379A2, WO 02/064594A2, WO 02/083622A2, WO 02/094842A2, WO 02/096426A1 , WO 02/101015A2, WO 02/103000A2, WO 03/008413A1, WO 03/016248A2, WO 03/020715A1 , WO 03/024899A2, WO 03/031431 A1 , WO 03/040103A1 , WO 03/053940A1 , WO 03/053941 A2, WO 03/063799A2, WO 03/079986A2, WO 03/080024A2, WO 03/082287A1 , WO 97/44467A1 , WO 99/01449A1 , and WO 99/58523A1), and immunomodulatory agents (
  • Patent No. 6,258,823 and everolimus and derivatives thereof (e.g., U.S. Patent No. 5,665,772).
  • Further representative examples of sirolimus analogues and derivatives include ABT-578 and those found in PCT Publication Nos.
  • biologically active agents which may be combined with implantable pump and sensor devices according to the invention include tyrosine kinase inhibitors, such as imantinib, ZK-222584, CGP-52411 , CGP-53716, NVP-AAK980-NX, CP-127374, CP-564959, PD-171026, PD- 173956, PD-180970, SU-0879, and SKI-606; MMP inhibitors such as nimesulide, PKF-241-466, PKF-242-484, CGS-27023A, SAR-943, primomastat, SC-77964, PNU-171829, AG-3433, PNU-142769, SU-5402, and dexlipotam; p38 MAP kinase inhibitors such as include CGH-2466 and PD-98-59; immunosuppressants such as argyrin B, macrocyclic lactone, ADZ-62-826,
  • cytokine inhibitors such as TNF-484A, PD-172084, CP-293121 , CP-353164, and PD- 168787; NFKB inhibitors, such as, AVE-0547, AVE-0545, and IPL-576092; HMGCoA reductase inhibitors, such as, pravestatin, atorvastatin, fluvastatin, dalvastatin, glenvastatin, pitavastatin, CP-83101 , U-20685; apoptosis antagonist (e.g., troloxamine, TCH-346 (N-methyl-N-propargyl-10-aminomethyl- dibenzo(b,f)oxepin); and caspase inhibitors (e.g., PF-5901 (benzenemethanol, alpha-pentyl-3-(2-quinolinyl)
  • the implantable pump and sensor devices may further include an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
  • an antibiotic e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir.
  • a polymeric composition comprising a fibrosis- inhibiting agent is combined with an agent that can modify metabolism of the agent in vivo to enhance efficacy of the fibrosis-inhibiting agent.
  • One class of therapeutic agents that can be used to alter drug metabolism includes agents capable of inhibiting oxidation of the anti-scarring agent by cytochrome P450 (CYP).
  • compositions include a fibrosis- inhibiting agent (e.g., The following agents are exemplary compounds: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus,
  • CYP inhibitors include flavones, azole antifungals, macrolide antibiotics, HIV protease inhibitors, and anti-sense oligomers.
  • Devices comprising a combination of a fibrosis-inhibiting agent and a CYP inhibitor may be used to treat a variety of proliferative conditions that can lead to undesired scarring of tissue, including intimal hyperplasia, surgical adhesions, and tumor growth.
  • a device incorporates or is coated on one aspect, portion or surface, portion or surface with a composition which inhibits fibrosis (and/or restenosis), as well as with a composition or compound which promotes or stimulates fibrosis on another aspect, portion or surface, portion or surface of the device.
  • Compounds that promote or stimulate fibrosis can be identified by, for example, the in vivo (animal) models provided in Examples 47-50.
  • agents that promote fibrosis include siik and other irritants (e.g., talc, wool (including animal wool, wood wool, and synthetic wool), talcum powder, copper, metallic beryllium (or its oxides), quartz dust, silica, crystalline silicates), polymers (e.g., polylysine, polyurethanes, poly(ethylene terephthalate), PTFE, poly(alkylcyanoacrylates), and poly(ethylene-co-vinylacetate); vinyl chloride and polymers of vinyl chloride; peptides with high lysine content; growth factors and inflammatory cytokines involved in angiogenesis, fibroblast migration, fibroblast proliferation, ECM synthesis and tissue remodeling, such as epidermal growth factor (EGF) family, transforming growth factor- ⁇ (TGF- ⁇ ), transforming growth factor- ⁇ (TGF- ⁇ -1 , TGF- ⁇ -2, TGF- ⁇ -3, platelet-derived growth factor (PDGF), fibroblast growth factor (acidic fibros
  • CTGF connective tissue growth factor
  • inflammatory microcrystals e.g., crystalline minerals such as crystalline silicates
  • bromocriptine methylsergide, methotrexate, chitosan, N-carboxybutyl chitosan, carbon tetrachloride, thioacetamide, fibrosin, ethanol, bleomycin, naturally occurring or synthetic peptides containing the Arg-Gly-Asp (RGD) sequence, generally at one or both termini (see, e.g., U.S. Patent No. 5,997,895), and tissue adhesives, such as cyanoacrylate and crosslinked poly(ethylene glycol) - methylated collagen compositions.
  • tissue adhesives such as cyanoacrylate and crosslinked poly(ethylene glycol) - methylated collagen compositions.
  • fibrosis-inducing agents include bone morphogenic proteins (e.g., BMP-2, BMP- 3, BMP-4, BMP-5, BMP-6 (VgM), BMP-7 (OP-1), BMP-8, BMP-9, BMP-10, BMP-11 , BMP-12, BMP-13, BMP-14, BMP-15, and BMP-16.
  • BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, and BMP-7 are of particular utility.
  • Bone morphogenic proteins are described, for example, in U.S. Patent Nos.
  • fibrosis-inducing agents include components of extracellular matrix (e.g., fibronectin, fibrin, fibrinogen, collagen (e.g., bovine collagen), including fibrillar and non-fibrillar collagen, adhesive glycoproteins, proteoglycans (e.g., heparin sulfate, chondroitin sulfate, dermatan sulfate), hyaluronan, secreted protein acidic and rich in cysteine (SPARC), thrombospondins, tenacin, and cell adhesion molecules (including integrins, vitronectin, fibronectin, laminin, hyaluronic acid, elastin, bitronectin), proteins found in basement membranes, and fibrosin) and inhibitors of matrix metalloproteinases, such as TIMPs (tissue inhibitors of matrix metalloproteinases) and synthetic TIMPs, such as, e.g., marimistat,
  • combretastatin A4 may be understood to refer to not only the common chemically available form of combretastatin, but analogues (e.g., combretastatin A2, A3, A5, A6, as noted above) and combretastatin conjugates.
  • analogues e.g., combretastatin A2, A3, A5, A6, as noted above
  • combretastatin conjugates e.g., combretastatin A2, A3, A5, A6, as noted above
  • the agents set forth above may be noted within the context of one class, many of the agents listed in fact have multiple biological activities. Further, more than one therapeutic agent may be utilized at a time (i.e., in combination), or delivered sequentially.
  • Drug dose can be calculated as a function of dose (i.e., amount) per unit area of the portion of the device being coated. Surface area can be measured or determined by methods known to one of ordinary skill in the art. Total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single systemic dose application.
  • the drug is released in effective concentrations for a period ranging from 1 - 90 days.
  • the fibrosis-inhibiting agents used alone or in combination, may be administered under the following dosing guidelines:
  • implantable sensors and pumps may be used in combination with a composition that includes an anti-scarring agent.
  • the total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 ⁇ g-10 ⁇ g, or 10 ⁇ g-10 mg, or 10 mg-250 mg, or 250 mg- 1000 mg, or 1000 mg-2500 mg.
  • the dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ⁇ g/mm 2 - 1 ⁇ g/mm 2 , or 1 ⁇ g/mm 2 - 10 ⁇ g/mm 2 , or 10 ⁇ g/mm 2 - 250 ⁇ g/mm 2 , 250 ⁇ g/mm 2 - 1000 ⁇ g/mm 2 , or 1000 ⁇ g/mm 2 - 2500 ⁇ g/mm 2 .
  • the total dose typically should not exceed 200 mg (range of 0.1 ⁇ g to 200 mg) and preferably 1 ⁇ g to 100 mg; dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 ; and minimum concentration of 10 '8 - 10 '4 M of agent should be maintained on the implant or barrier surface.
  • the total dose typically should not to exceed 500 mg (range of 1.0 ⁇ g to 500 mg) and preferably 1 ⁇ g to 200 mg; dose per unit area of 0.01 ⁇ g - 200 ⁇ g per mm 2 , preferably 0.1 ⁇ g/mm 2 - 40 ⁇ g/mm 2 .
  • the total dose typically should not exceed 1000 mg (range of 0.1 ⁇ g to 1000 mg), preferably 1 ⁇ g to 500 mg; dose per unit area of 0.01 ⁇ g - 500 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 100 ⁇ g/mm 2 . It may be apparent to one of skill in the art that potentially any anti- fibrosis agent described above may be utilized alone, or in combination, in the practice of this embodiment.
  • the present invention provides an implantable sensor or pump that contains an anti-fibrosing agent listed below in a dosage as set forth above: 1) an anti-fibrotic agent that inhibits cell regeneration, 2) an anti-fibrotic agent that inhibits angiogenesis, 3) an anti-fibrotic agent that inhibits fibroblast migration, 4) an anti-fibrotic agent that inhibits fibroblast proliferation, 5) an anti-fibrotic agent that inhibits deposition of extracellular matrix, 6) an anti- fibrotic agent inhibits tissue remodeling, 7) an adensosine A2A receptor antagonist, 8) an AKT inhibitor, 9) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No.
  • an alpha 4 integrin antagonist an alpha 4 integrin antagonist
  • an alpha 7 nicotinic receptor agonist 12) an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceutical
  • a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof, 39) an endotoxin antagonist, 40) an epothilone and tubulin binder, 41) an estrogen receptor antagonist, 42) an FGF inhibitor, 43) a farnexyl transferase inhibitor, 44) a famesyltransferase inhibitor selected from the group of A-197574 (Abbott),
  • an FLT-3 kinase inhibitor 46a) an FGF receptor kinase inhibitor, 47) a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No.
  • a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17- dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1 l ,4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No.
  • an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No.
  • an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No.
  • 334476- 64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof, 75) an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No.
  • INDRA OXiGENE
  • IPL- 576092 CAS No. 137571-30-3
  • Inflazyme NFKB decoy
  • NFKB decoy oligo NFKB decoy oligo
  • S5 F005 from Fulcrum Pharmaceuticals
  • P61 Physicaltopharm
  • R-flurbiprofen CAS No.
  • a palmitoyl-protein thioesterase inhibitor 80) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No.
  • a peroxisome proliferators-activated receptor agonist selected from the group consisting of (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No.
  • 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No.
  • MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof, 82) a phosphatase inhibitor, 83) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No.
  • PDE phosphodiesterase
  • a phosphodiesterase III inhibitor enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof
  • a PKC inhibitor 85) a platelet activating factor antagonist, 86) a platelet-derived growth factor receptor kinase inhibitor, 87) a prolyl hydroxylase inhibitor, 88) a polymorphonuclear neutrophil inhibitor, 89) a protein kinase B inhibitor, 90) a protein kinase C stimulant, 91) a purine nucleoside analogue, 92) a purinoreceptor P2X antagonist, 93) a Raf kinase inhibitor, 94) a reversible inhibitor of ErbB1 and ErbB2, 95) a ribonucleoside triphosphate reductase inhibitor, 96
  • TNF ⁇ antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No.
  • TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No.
  • brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No.
  • H3 receptor antagonists e.g., thioperamide and thioperamide maleate salt
  • anti-histamines e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones
  • 131 an alpha adrenergic receptor antagonist
  • 132) an anti-psychotic compound
  • 133) a CaM kinase Il inhibitor
  • 134) a G protein agonist 135) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No.
  • a DNA topoisomerase inhibitor selected from the group consisting of ⁇ -lapachone (CAS No. 4707-32- 8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof, 138) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No.
  • a D2 dopamine receptor antagonist 140) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, 141) a dopamine antagonist, an anesthetic compound, 142) a clotting factor, 143) a lysyl hydrolase inhibitor, 144) a muscarinic receptor inhibitor, 145) a superoxide anion generator, 146) a steroid, 147) an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No.
  • temozolomide temozolomide
  • procarbazine HCI and an analogue or derivative thereof
  • 157) a DNA methylation inhibitor 158) a NSAID agent, 159) a peptidylglycine alpha- hydroxylating monooxygenase inhibitor, 160) an MEK1/MEK 2 inhibitor, 161) a NO synthase inhibitor, 162) a retinoic acid receptor antagonist selected from isotretinoin (CAS No.
  • an ACE inhibitor 164) a glycosylation inhibitor, 165) an intracellular calcium influx inhibitor, 166) an anti-emetic agent, 167) an acetylcholinesterase inhibitor, 168) an ALK-5 receptor antagonist, 169) a RAR/RXT antagonist, 170) an elF-2a inhibitor, 171) an S-adenosyl-L-homocysteine hydrolase inhibitor, 172) an estrogen agonist, 173) a serotonin receptor inhibitor, 174) an anti-thrombotic agent, 175) a tryptase inhibitor, 176) a pesticide, 177) a bone mineralization promoter, 178) a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, 179) an anti-inflammatory compound, 180) a DNA methylation promoter, 181) an anti-spasmodic agent, 182) a protein synthesis inhibitor, 183) an ⁇ -glucosidase inhibitor
  • Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 .
  • mTOR inhibitors including AP-23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 . Minimum concentration of 10 ' 8 - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • TNF alpha antagonists including etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 . Minimum concentration of 10 ⁇ - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • Minimum concentration of 10 '8 - 10 '4 M of agent is to be maintained on the implant or barrier surface.
  • NF Kappa B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 ⁇ g to 200 mg); preferred 1 ⁇ g to 100 mg. Dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferred dose of 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 . Minimum concentration of 10 ⁇ - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • Minimum concentration of 10 ⁇ - 10 "4 M of agent is to be maintained on the implant or barrier surface.
  • anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC 50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus.
  • selected assays described herein such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus.
  • the total dose typically should not exceed 200 mg (range of 0.1 ⁇ g to 200 mg) and preferably 1 ⁇ g to 100 mg; dose per unit area of 0.01 ⁇ g - 100 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 20 ⁇ g/mm 2 ; and minimum concentration of 10 ⁇ - 10 "4 M of agent should be maintained on the implant or barrier surface.
  • agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC 5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate.
  • the total dose typically should not to exceed 500 mg (range of 1.0 ⁇ g to 500 mg) and preferably 1 ⁇ g to 200 mg; dose per unit area of 0.01 ⁇ g - 200 ⁇ g per mm 2 , preferably 0.1 ⁇ g/mm 2 - 40 ⁇ g/mm 2 ; and minimum concentration of 10 ⁇ - 10 "4 M of agent should be maintained on the implant or barrier surface.
  • agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC 5 0 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
  • the total dose typically should not exceed 1000 mg (range of 0.1 ⁇ g to 1000 mg), preferably 1 ⁇ g to 500 mg; dose per unit area of 0.01 ⁇ g - 500 ⁇ g per mm 2 ; preferably 0.1 ⁇ g/mm 2 - 100 ⁇ g/mm 2 ; and minimum concentration of 10 ⁇ - 10 "4 M of agent should to be maintained on the implant or barrier surface.
  • fibrosis is inhibited by local, regional or systemic release of specific pharmacological agents that become localized to the tissue adjacent to the device or implant.
  • implantable sensors or implantable pumps where the occurrence of a f ibrotic reaction will adversely affect the functioning of the device or the biological problem for which the device was implanted or used.
  • fibrotic encapsulation of the device slows, impairs, or interrupts detection (sensors) or drug delivery (pumps) to/from the device to/from the tissue. This can cause the device to function suboptimally or not at all, negatively affect disease management, and/or shorten the lifespan of the device.
  • sensors sensors
  • drug delivery umps
  • Medical devices or implants of the present invention are coated with, or adapted to release an agent which inhibits fibrosis on the surface of, or around, the implantable sensor and/or implantable pump.
  • the present invention provides implantable sensors and implantable pumps that include an anti-scarring agent or a composition that includes an anti-scarring agent such that the overgrowth of fibrous or granulation tissue is inhibited or reduced.
  • Methods for incorporating fibrosis-inhibiting compositions onto or into implantable sensors and implantable pumps include: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis- inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) by inserting the device into a sleeve or mesh which is comprised of, or coated with, a fibrosis-inhibiting composition, (f) constructing the device itself (or a portion of the device such as the detector, drug
  • the coating process can be performed in such a manner as to coat all or parts (such as the sensor or the drug delivery catheter/port) of the entire device with the fibrosis-inhibiting composition.
  • the fibrosis-inhibiting agent can be mixed with the materials that are used to make the implantable sensor or implantable pump such that the fibrosis-inhibiting agent is incorporated into the final product.
  • a medical device may be prepared which has a coating, where the coating is, e.g., uniform, non-uniform, continuous, discontinuous, or patterned.
  • an implantable sensor or drug delivery/catheter/port device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug (i.e., one or more fibrosis-inhibiting agents).
  • the reservoirs may be formed from divets in the device surface or micropores or channels in the device body.
  • the reservoirs are formed from voids in the structure of the device.
  • the reservoirs may house a single type of drug (e.g., fibrosis-inhibiting agent) or more than one type of drug (e.g., a fibrosis-inhibiting agent and an anti-infective agent).
  • the drug(s) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs.
  • a carrier e.g., a polymeric or non-polymeric material
  • the filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier.
  • the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount
  • the multi- layered carrier may further include a barrier layer that prevents release of the drug(s).
  • the barrier layer can be used, for example, to control the direction that the drug elutes from the void.
  • the coating of the medical device may directly contact the implantable device, or it may indirectly contact the device when there is something, e.g., a polymer layer, that is interposed between the device and the coating that contains the fibrosis-inhibiting agent.
  • the fibrosis-inhibiting agent can be applied directly or indirectly to the tissue adjacent to the implantable sensors and implantable pump (preferably near the interface of the tissue and the detector, drug delivery catheter and/or drug delivery port).
  • the fibrosis-inhibiting agent with or without a polymeric, non-polymeric, or secondary carrier: (a) to the device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) prior to, immediately prior to, or during, implantation of the implantable sensors and implantable pump; (c) to the surface of the device and/or the tissue surrounding the implanted pump or sensor (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after implantation; (d) by topical application of the anti-fibrosis agent into the anatomical space where the implantable sensors and implantable pump will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microe
  • Combination therapies ⁇ i.e., combinations of therapeutic agents and combinations with antithrombotic, antiplatelet and/or anti-infective agents
  • the anti-fibrosing agent can be coated onto the entire device or a portion of the device.
  • the agent is present as part of a coating on a surface of the implantable sensor or implantable pump.
  • the coating may partially cover or may completely cover the surface of the implantable sensor or implantable pump. Further, the coating may directly or indirectly contact the implantable sensor or implantable pump.
  • the Implantable sensor or implantable pump may be coated with a first coating and then coated with a second coating that includes the anti- scarring agent.
  • Implantable sensors and implantable pumps may be coated using a variety of coating methods, including by dipping, spraying, painting, by vacuum deposition, or by any other method known to those of ordinary skill in the art.
  • the anti-fibrosing agent can be coated onto the appropriate implantable sensors and implantable pumps using the polymeric coatings described above.
  • the coating compositions and methods described above there are various other coating compositions and methods that are known in the art. Representative examples of these coating compositions and methods are described in U.S. Patent. Nos.
  • the biologically active fibrosis-inhibiting agent can be delivered with non-polymeric agents.
  • non-polymeric agents can include sucrose derivatives (e.g., sucrose acetate isobutyrate, sucrose oleate), sterols such as cholesterol, stigmasterol, beta- sitosterol, and estradiol; cholesteryl esters such as cholesteryl stearate; Ci 2 - C 24 fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, and lignoceric acid; C 18 -C 36 mono-, di- and triacylglycerides such as glyceryl monooleate, glyceryl monolinoleate, glyceryl monolaurate, glyceryl monodocosanoate, glyceryl monomyristate, glyceryl monodicenoate, glyceryl dipalmitate, glyceryl didocosanoate, glyceryl dimyristate, glyceryl
  • the fibrosis-inhibiting agent may be delivered as a solution.
  • the fibrosis-inhibiting agent can be incorporated directly into the solution to provide a homogeneous solution or dispersion.
  • the solution is an aqueous solution.
  • the aqueous solution may futher include buffer salts, as well as viscosity modifying agents (e.g., hyaluronic acid, alginates, CMC, and the like).
  • the solution can include a biocompatible solvent, such as ethanol, DMSO, glycerol, PEG-200, PEG-300 or NMP.
  • the fibrosis-inhibiting agent can further comprise a secondary carrier.
  • the secondary carrier can be in the form of microspheres (e.g., PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate), nanospheres (e.g., PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), liposomes, emulsions, microemulsions, micelles (e.g., SDS, block copolymers of the form X-Y, X-Y-X or Y-X-Y where X is a poly(alkylene oxide) or alkyl ether thereof and Y is a polyester (e.g., PLGA, PLLA, PDLLA, PCL polydioxanone)), zeolites or cyclodextrins.
  • microspheres e.g.

Landscapes

  • Health & Medical Sciences (AREA)
  • Vascular Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Anesthesiology (AREA)
  • Biomedical Technology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Infusion, Injection, And Reservoir Apparatuses (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Pumps and sensors for contact with tissue are used in combination with an anti-scarring agent or a composition that comprises an anti-scarring agent to inhibit scarring that may otherwise occur when the pumps and sensors are implanted within an animal.

Description

IMPLANTABLE SENSORS AND PUMPS, ANTI-SCARRING AGENTS, AND
THERAPEUTIC COMPOSITIONS
BACKGROUND OF THE INVENTION
Field of the Invention The present invention relates generally to implantable sensors, drug-delivery devices and drug-delivery pump, and more specifically, to compositions and methods for preparing and using such devices to make them resistant to overgrowth by inflammatory and fibrous scar tissue.
Description of the Related Art Implantable drug delivery devices and pumps are a means to provide prolonged, site-specific release of a therapeutic agent for the management of a variety of medical conditions. Drug delivery implants and pumps are generally utilized when a localized pharmaceutical impact is desired (i.e., the condition affects only a specific region) or when systemic delivery of the agent is inefficient or ineffective and leads toxicity, severe side effects, inactivation of the drug prior to reaching the target tissue, poor symptom/disease control, and/or addiction to the medication. Implantable pumps can also deliver systemic drug levels in a constant, regulated manner for extended periods and help patients avoid the "peaks and valleys" of blood-level drug concentrations associated with intermittent systemic dosing. For many patients this can lead to better symptom control (the dosage can often be titrated to the severity of the symptoms), superior disease management (particularly for insulin delivery in diabetics), and lower drug requirements (particularly for pain medications). Innumerable drug delivery devices, implants and pumps have been developed for an array of specific medical conditions and the particular construction and delivery mechanism of the device depends on the particular treatment. For example, drug delivery implants and pumps have been used in a variety of clinical applications, including programmable insulin pumps for the treatment of diabetes, intrathecal (in the spine) pumps to administer narcotics (e.g., morphine, fentanyl) for the relief of pain (e.g., cancer, back problems, HIV, post-surgery), local and systemic delivery of chemotherapy for the treatment of cancer (e.g., hepatic artery 5-FU infusion for liver tumors), medications for the treatment of cardiac conditions (e.g., antiarrhythmic drugs for cardiac rhythm abnormalities), intrathecal delivery of anti- spasmotic drugs (e.g., baclofen) for spasticity in neurological disorders (e.g., Multiple Sclerosis, spinal cord injuries, brain injury, cerebral palsy), or local/regional antibiotics for infection management (e.g., osteomyelitis, septic arthritis).
Typically, most drug delivery pumps are implanted subcutaneously (under the skin in an easy to access, but discrete location) and consist of a pump unit with a drug reservoir and a flexible catheter through which the drug is delivered to the target tissue. The pump stores and releases prescribed amounts of medication via the catheter to achieve therapeutic drug levels either locally or systemically (depending upon the application). The center of the pump has a self-sealing access port covered by a septum such that a needle can be inserted percutaneously (through both the skin and the septum) to refill the pump with medication as required. There are generally two types of implantable drug delivery pumps. Constant-rate pumps are usually powered by gas and are designed to dispense drugs under pressure as a continual dosage at a preprogrammed, constant rate. The amount and rate of drug flow are regulated by the length of the catheter used, temperature and altitude, and they are best when unchanging, long-term drug delivery is required. Although limited, these pumps have the advantage of being simple, having few moving parts, not requiring battery power and possessing a longer lifespan. Programmable-rate pumps utilize a battery-powered pump and a constant pressure reservoir to deliver drugs on a periodic basis in a manner that can be programmed by the physician or the patient. For the programmable infusion device, the drug may be delivered in small, discrete doses based on a programmed regimen which can be altered according to an individual's clinical response. Programmable drug delivery pumps may be in communication with an external transmitter which programs the prescribed dosing regimen, including the rate, time and amount of each dose, via low-frequency waves that are transmitted through the skin. Programmable-rate pumps are more widely used and provide superior dosimetry, but because of their complexity, they require more maintenance and have a shorter lifespan.
The clinical function of an implantable drug delivery device or pump depends upon the device, particularly the catheter, being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum) and not becoming encapsulated or obstructed by scar tissue. Unfortunately, in many instances when these devices are implanted in the body, they are subject to a "foreign body" response from the surrounding host tissues. The body recognizes the implanted device as foreign, which triggers an inflammatory response followed by encapsulation of the implant with fibrous connective tissue. Scarring (i.e., fibrosis) can also result from trauma to the anatomical structures and tissue surrounding the implant during implantation of the device. Lastly, fibrous encapsulation of the device can occur even after a successful implantation if the device is manipulated (some patients continuously "fiddle" with a subcutaneous implant) or irritated by the daily activities of the patient. For drug delivery pumps, the catheter tip or lumen may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Alternatively, the catheter can become encapsulated by scar (i.e., the body "walls off' the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement from the catheter to the tissues on the other side of the capsule). Either of these developments may lead to inefficient or incomplete drug flow to the desired target tissues or organs (and loss of clinical benefit), while the second can also lead to local drug accumulation (in the capsule) and additional clinical complications (e.g., local drug toxicity; drug sequestration followed by sudden "dumping" of large amounts of drug into the surrounding tissues). Additionally, the tissue surrounding the implantable pump or catheter can be inadvertently damaged from the inflammatory foreign body response leading to loss of function and/or tissue damage (e.g., scar tissue in the spinal canal causing pain or obstructing the flow of cerebrospinal fluid). A device that is frequently (but not always) used in association with a drug delivery pump is an implantable sensor device. An implantable sensor is a device used to detect changes in body function and/or levels of key physiological metabolites, chemistry, hormones or biological factors. Implantable sensors may be used to sense a variety of physical and/or physiological properties, including, but not limited to, optical, mechanical, chemical, electrochemical, temperature, strain, pressure, magnetism, acceleration, ionizing radiation, acoustic wave or chemical changes. Often sensor technology is combined with implantable drug delivery pumps such that the sensor receives a signal and then, in turn, uses this information to modulate the release kinetics of a drug. The most widely pursued application of this technology is the production of a closed-loop "artificial pancreas" which can continuously detect blood glucose levels (through an implanted sensor) and provide feedback to an implantable pump to modulate the administration of insulin to a diabetic patient. Other representative examples of implantable sensors include, blood/tissue glucose monitors, electrolyte sensors, blood constituent sensors, temperature sensors, pH sensors, optical sensors, amperometric sensors, pressure sensors, biosensors, sensing transponders, strain sensors, activity sensors and magnetoresistive sensors. Much like the problem facing drug delivery pumps described above, proper clinical functioning of an implanted sensor is dependent upon intimate anatomical contact with the target tissues and/or body fluids. Scarring around the implanted device may degrade the electrical components and characteristics of the device-tissue interface, and the device may fail to function properly. For example, when a "foreign body" response occurs and the implanted sensor becomes encapsulated by scar (Ae., the body "walls off' the sensor with fibrous tissue), the sensor receives inaccurate biological information. If the sensor is detecting conditions inside the capsule, and these conditions are not consistent with those outside the capsule (which is frequently the case), it will produce inaccurate readings. Similarly if the scar tissue alters the flow of physical or chemical information to the detection mechanism of the sensor, the information it processes will not be reflective of those present in the target tissue.
BRIEF SUMMARY OF THE INVENTION
Briefly stated, the present invention discloses pharmaceutical agents which inhibit one or more aspects of the production of excessive fibrous (scar) tissue. In one aspect, the present invention provides compositions for delivery of selected therapeutic agents via medical devices or implants containing sensors or drug delivery pumps, as well as methods for making and using these implants and devices. Compositions and methods are described for coating sensors or pumps with drug-delivery compositions such that the pharmaceutical agent is delivered in therapeutic levels over a period sufficient to prevent the drug delivery catheter and/or the implanted sensor from being encapsulated in fibrous tissue to improve and/or prolong device function. Alternatively, locally administered compositions (e.g., topicals, injectables, liquids, gels, sprays, microspheres, pastes, wafers) containing an inhibitor of fibrosis are described that can be applied to the tissue adjacent to the implanted pump (particularly the delivery catheter) and/or the implanted sensor, such that the fibrosis-inhibitor is delivered in therapeutic levels over a period sufficient to prevent the delivery catheter or sensor from being occluded or encapsulated by fibrous tissue. And finally, numerous specific implantable pumps, sensors and combined devices are described that produce superior clinical results as a result of being coated with agents that reduce excessive scarring and fibrous tissue accumulation as well as other related advantages.
Within one aspect of the invention, drug-coated or drug- impregnated implants and medical devices are provided which reduce fibrosis in the tissue surrounding the implanted drug delivery pump or sensor, or inhibit scar development on the device/implant surface (particularly the drug delivery catheter lumen and the sensor surface), thus enhancing the efficacy of the procedure. For example, fibrous tissue can reduce or obstruct the flow of therapeutic agents from the catheter to the target tissue, or prevent the implanted sensor from detecting accurate readings. Within various embodiments, fibrosis is inhibited by local or systemic release of specific pharmacological agents that become localized to the tissue adjacent to the implanted device.
The repair of tissues following a mechanical or surgical intervention, such as the implantation of a pump or sensor, involves two distinct processes: (1) regeneration (the replacement of injured cells by cells of the same type and (2) fibrosis (the replacement of injured cells by connective tissue). There are several general components to the process of fibrosis (or scarring) including: infiltration of inflammatory cells and the inflammatory response, migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), formation of new blood vessels (angiogenesis), and remodeling (maturation and organization of the fibrous tissue). As utilized herein, "inhibits (reduces) fibrosis" may be understood to refer to agents or compositions which decrease or limit the formation of fibrous tissue (i.e., by reducing or inhibiting one or more of the processes of inflammation, connective tissue cell migration or proliferation, ECM production, angiogenesis, and/or remodeling). In addition, numerous therapeutic agents described in this invention will have the additional benefit of also reducing tissue regeneration where appropriate.
Within certain embodiments of the invention, an implant or device (e.g., a sensor or pump) is adapted to release an agent that inhibits fibrosis through one or more of the mechanisms cited herein. Within certain other embodiments of the invention, an implant or device contains an agent that while remaining associated with the implant or device, inhibits fibrosis between the implant or device and the tissue where the implant or device is placed by direct contact between the agent and the tissue surrounding the implant or device. Within related aspects of the present invention, implanted pumps and sensors are provided comprising an implant or device, wherein the implant or device releases an agent which inhibits fibrosis in vivo. "Release of an agent" refers to any statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the implant/device and/or remains active on the surface of (or within) the device/implant. Within yet other aspects of the present invention, methods are provided for manufacturing a medical device or implant, comprising the step of coating (e.g., spraying, dipping, wrapping, or administering drug through) a medical device or implant. Additionally, the implant or medical device can be constructed so that the device itself is comprised of materials which inhibit fibrosis in or around the implant. A wide variety of implantable pumps and sensors may be utilized within the context of the present invention, depending on the site and nature of treatment desired. Within various embodiments of the invention, the implanted pump or sensor is further coated with a composition or compound, which delays the onset of activity of the fibrosis-inhibiting agent for a period of time after implantation. Representative examples of such agents include heparin, PLGA/MePEG, PLA, and polyethylene glycol. Within further embodiments, the fibrosis-inhibiting implant or device is activated before, during, or after deployment (e.g., an inactive agent on the device is first activated to one that reduces or inhibits an in vivo fibrotic reaction).
Within various embodiments of the invention, the tissue surrounding the implanted pump (particularly the drug delivery catheter) and/or sensor is treated with a composition or compound that contains an inhibitor of fibrosis. Locally administered compositions (e.g., topicals, injectables, liquids, gels, sprays, microspheres, pastes, wafers) or compounds containing an inhibitor of fibrosis are described that can be applied to the surface of, or infiltrated into, the tissue adjacent to the pump or sensor, such that the pharmaceutical agent is delivered in therapeutic levels over a period sufficient to prevent the drug delivery catheter and/or sensor from being obstructed or encapsulated by fibrous tissue. This can be done in lieu of coating the device or implant with a fibrosis-inhibitor, or done in addition to coating the device or implant with a fibrosis-inhibitor. The local administration of the fibrosis-inhibiting agent can occur prior to, during, or after implantation of the pump or sensor itself.
Within various embodiments of the invention, an implanted pump or sensor is coated on one aspect, portion or surface with a composition which inhibits fibrosis, as well as being coated with a composition or compound which promotes scarring on another aspect, portion or surface of the device (i.e., to affix the body of the device into a particular anatomical space). Representative examples of agents that promote fibrosis and scarring include silk, silica, crystalline silicates, bleomycin, quartz dust, neomycin, talc, metallic beryllium and oxides thereof, retinoic acid compounds, copper, leptin, growth factors, a component of extracellular matrix; fibronectin, collagen, fibrin, or fibrinogen, polylysine, poly(ethylene-co-vinylacetate), chitosan, N-carboxybutylchitosan, and RGD proteins; vinyl chloride or a polymer of vinyl chloride; an adhesive selected from the group consisting of cyanoacrylates and crosslinked poly(ethylene glycol) - methylated collagen; an inflammatory cytokine (e.g., TGFβ, PDGF, VEGF, bFGF, TNFα, NGF, GM-CSF, IGF-1 , IL-1 , IL-1-β, IL-8, IL- 6, and growth hormone); connective tissue growth factor (CTGF) as well as analogues and derivatives thereof.
Also provided by the present invention are methods for treating patients undergoing surgical, endoscopic or minimally invasive therapies where an implanted pump or sensor is placed as part of the procedure. As utilized herein, it may be understood that "inhibits fibrosis" refers to a statistically significant decrease in the amount of scar tissue in or around the device or an improvement in the interface between the implant (catheter and/or sensor) and the tissue, which may or may not lead to a permanent prohibition of any complications or failures of the device/implant. The pharmaceutical agents and compositions are utilized to create novel drug-coated implants and medical devices that reduce the foreign body response to implantation and limit the growth of reactive tissue on the surface of, into, or around the device, such that performance is enhanced. Implantable pumps and sensors coated with selected pharmaceutical agents designed to prevent scar tissue overgrowth and improve electrical conduction can offer significant clinical advantages over uncoated devices.
For example, in one aspect the present invention is directed to implantable pumps and sensors that comprise a medical implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti- scarring agent. The agent is present so as to inhibit scarring that may otherwise occur when the implant is placed within an animal. In another aspect the present invention is directed to methods wherein both an implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti-scarring agent, are placed into an animal, and the agent inhibits scarring that may otherwise occur. These and other aspects of the invention are summarized below.
Thus, in various independent aspects, the present invention provides a device, comprising an implantable pump and/or sensor and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring. These and other devices are described in more detail herein.
In additional aspects, for each of the aforementioned devices combined with each of the agents described herein, it is, for each combination, independently disclosed that the agent may be present in a composition along with a polymer. In one embodiment of this aspect, the polymer is biodegradable. In another embodiment of this aspect, the polymer is nonbiodegradable. Other features and characteristics of the polymer, which may serve to describe the present invention for every combination of device and agent described above, are set forth in greater detail herein.
In addition to devices, the present invention also provides methods. For example, in additional aspects of the present invention, for each of the aforementioned devices, and for each of the aforementioned combinations of the devices with the anti-scarring agents, the present invention provides methods whereby a specified device is implanted into an animal, and a specified agent associated with the device inhibits scarring that may otherwise occur. Each of the devices identified herein may be a "specified device", and each of the anti-scarring agents identified herein may be an "anti- scarring agent", where the present invention provides, in independent embodiments, for each possible combination of the device and the agent.
The agent may be associated with the device prior to the device being placed within the animal. For example, the agent (or composition comprising the agent) may be coated onto an implant, and the resulting device then placed within the animal. In addition, or alternatively, the agent may be independently placed within the animal in the vicinity of where the device is to be, or is being, placed within the animal. For example, the agent may be sprayed or otherwise placed onto, adjacent to, and/or within the tissue that will be contacting the medical implant or may otherwise undergo scarring. To this end, the present invention provides placing an implantable pump and/or sensor and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
In additional aspects, for each of the aforementioned methods used in combination with each of the agents described herein, it is, for each combination, independently disclosed that the agent may be present in a composition along with a polymer. In one embodiment of this aspect, the polymer is biodegradable. In another embodiment of this aspect, the polymer is non-biodegradable. Other features and characteristics of the polymer, which may serve to describe the present invention for every combination of device and agent described above, are set forth in greater detail herein.
In each of the aforementioned devices, compositions, methods of making the aforementioned devices or compositions, and methods of using the aforementioned devices or compostions, the present invention provides that the anti-fibrotic agent may be one or more of the following: 1) an anti-fibrotic agent that inhibits cell regeneration, 2) an anti-fibrotic agent that inhibits angiogenesis, 3) an anti-fibrotic agent that inhibits fibroblast migration, 4) an anti-fibrotic agent that inhibits fibroblast proliferation, 5) an anti-fibrotic agent that inhibits deposition of extracellular matrix, 6) an anti-fibrotic agent inhibits tissue remodeling, 7) an adensosine A2A receptor antagonist, 8) an AKT inhibitor, 9) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is
Pharmaprojects No. 5754 (Merck KgaA), 10) an alpha 4 integrin antagonist, 11) an alpha 7 nicotinic receptor agonist, 12) an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC- 4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists
(Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT- 116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV- 6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB)1 JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof, 13) an apoptosis antagonist, 14) an apoptosis activator, 15) a beta 1 integrin antagonist, 16) a beta tubulin inhibitor, 17) a blocker of enzyme production in Hepatitis C, 18) a Bruton's tyrosine kinase inhibitor, 19) a calcineurin inhibitor, 20) a caspase 3 inhibitor, 21) a CC chemokine receptor antagonist, 22) a cell cycle inhibitor selected from the group consisting of SNS- 595 (Sunesis), synthadotin, KRX-0403, homoharringtonine, and an analogue or derivative thereof, 23) a cathepsin B inhibitor, 24) a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof, 25) a cathepsin L inhibitor, 26) a CD40 antagonist, 27) a chemokine receptor agonist, 28) a chymase inhibitor, 29) a collagenase antagonist, 30) a CXCR antagonist, 31) a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof, 32) a cyclooxygenase 1 inhibitor, 33) a DHFR inhibitor, 34) a dual integrin inhibitor, 35) an elastase inhibitor, 36) an elongation factor-1 alpha inhibitor, 37) an endothelial growth factor antagonist, 38) an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No. 125697-92-9), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof, 39) an endotoxin antagonist, 40) an epothilone and tubulin binder, 41) an estrogen receptor antagonist, 42) an FGF inhibitor, 43) a famexyl transferase inhibitor, 44) a farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from
Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof, 45) an FLT-3 kinase inhibitor, 46a) an FGF receptor kinase inhibitor, 47) a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, and an analogue or derivative thereof, 48) a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17- dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No. 12772-57-5), and an analogue or derivative thereof, 49) a histone deacetylase inhibitor, 50) an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No. 143201-11-0), and an analogue or derivative thereof, 51) an ICAM inhibitor, 52) an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma- Lerads), and an analogue or derivative thereof, 53) an IL-2 inhibitor, 54) an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof, 55) an IMPDH (inosine monophosphate), 56) an integrin antagonist, 57) an interleukin antagonist, 58) an inhibitor of type III receptor tyrosine kinase, 59) an irreversible inhibitor of enzyme methionine aminopeptidase type 2, 60) an isozyme selective delta protein kinase C inhibitor, 61) a JAK3 enzyme inhibitor, 62) a JNK inhibitor, 63) a kinase inhibitor, 64) a kinesin antagonist, 65) a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spagiumic acid (ZY-15106) (CAS No.3106-85- 2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof, 66) a MAP kinase inhibitor, 67) a matrix metalloproteinase inhibitor, 68) an MCP-CCR2 inhibitor, 69) an mTOR inhibitor, 70) an mTOR kinase inhibitor,71) a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gnMf (GNI), huC242-DM4, huMy9-6-DM1 (immunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, dolastatin 15 (CAS No. 123884-00-4), vincamine, and an analogue or derivative thereof, 72) an MIF inhibitor, 73) an MMP inhibitor, 74) a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911 -02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476- 64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof, 75) an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), Bay 11-7085, and an analogue or derivative thereof, 76) a nitric oxide agonist, 77) an ornithine decarboxylase inhibitor, 78) a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol- Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74-6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof, 79) a palmitoyl-protein thioesterase inhibitor, 80) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof, 81) a peroxisome proliferators-activated receptor agonist selected from the group consisting of (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L- 165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof, 82) a phosphatase inhibitor, 83) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66- 5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof, 84) a PKC inhibitor, 85) a platelet activating factor antagonist, 86) a platelet-derived growth factor receptor kinase inhibitor, 87) a prolyi hydroxylase inhibitor, 88) a polymorphonuclear neutrophil inhibitor, 89) a protein kinase B inhibitor, 90) a protein kinase C stimulant, 91) a purine nucleoside analogue, 92) a purinoreceptor P2X antagonist, 93) a Raf kinase inhibitor, 94) a reversible inhibitor of ErbB1 and ErbB2, 95) a ribonucleoside triphosphate reductase inhibitor, 96) an SDF-1 antagonist, 97) a sheddase inhibitor, 98) an SRC inhibitor, 99) a stromelysin inhibitor, 100) an Syk kinase inhibitor, 101) a telomerase inhibitor, 102) a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof, 103) a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57-7/188913- 58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130- 73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum
Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof, 104) a tumor necrosis factor antagonist, 105) a Toll receptor inhibitor, 106) a tubulin antagonist, 107) a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlII MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD- 2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), herbimycin A, and an analogue or derivative thereof, 108) a VEGF inhibitor, 109) a vitamin D receptor agonist, 110) ZD-6474 (an angiogenesis inhibitor), 111) AP-23573 (an mTOR inhibitor), 112) synthadotin (a tubulin antagonist), 113) S-0885 (a collagenase inhibitor), 114) aplidine (an elongation factor- 1 alpha inhibitor), 115) ixabepilone (an epithilone), 116) IDN-5390 (an angiogenesis inhibitor and an FGF inhibitor), 117) SB-2723005 (an angiogenesis inhibitor), 118) ABT-518 (an angiogenesis inhibitor), 119) combretastatin (an angiogenesis inhibitor), 120) anecortave acetate (an angiogenesis inhibitor), 121) SB-715992 (a kinesin antagonist), 122) temsirolimus (an mTOR inhibitor), and 123) adalimumab (a TNFα antagonist), 124) erucylphosphocholine (an ATK inhibitor), 125) alphastatin (an angiogenesis inhibitor), 126) bortezomib (an NF Kappa B inhibitor), 127) etanercept (a TNFα antagonist and TACE inhibitor), 128) humicade (a TNFα inhibitor), and 129) gefitinib (a tyrosine kinase inhibitor), 130) a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidϊne (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones), 131) an alpha adrenergic receptor antagonist, 132) an anti-psychotic compound, 133) a CaM kinase Il inhibitor, 134) a G protein agonist, 135) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof, 136) an anti-microbial agent, 137) a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32- 8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof, 138) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof, 139) a D2 dopamine receptor antagonist, 140) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, 141) a dopamine antagonist, an anesthetic compound, 142) a clotting factor, 143) a lysyl hydrolase inhibitor, 144) a muscarinic receptor inhibitor, 145) a superoxide anion generator, 146) a steroid, 147) an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514, spermine tetrahydrochloride, NG-methyl-L- arginine acetate salt, galardin, and an analogue or derivative thereof, 148) a diuretic, 149) an anti-coagulant, 150) a cyclic GMP agonist, 151) an adenylate cyclase agonist, 152) an antioxidant, 153) a nitric oxide synthase inhibitor, 154) an antineoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof, 155) a DNA synthesis inhibitor, 156) a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCl, and an analogue or derivative thereof, 157) a DNA methylation inhibitor, 158) a NSAID agent, 159) a peptidylglycine alpha- hydroxylating monooxygenase inhibitor, 160) an MEK1/MEK 2 inhibitor, 161) a NO synthase inhibitor, 162) a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof, 163) an ACE inhibitor, 164) a glycosylation inhibitor, 165) an intracellular calcium influx inhibitor, 166) an anti-emetic agent, 167) an acetylcholinesterase inhibitor, 168) an ALK-5 receptor antagonist, 169) a RAR/RXT antagonist, 170) an elF-2a inhibitor, 171) an S-adenosyl-L-homocysteine hydrolase inhibitor, 172) an estrogen agonist, 173) a serotonin receptor inhibitor, 174) an antithrombotic agent, 175) a tryptase inhibitor, 176) a pesticide, 177) a bone mineralization promoter, 178) a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, 179) an anti-inflammatory compound, 180) a DNA methylation promoter, 181) an anti-spasmodic agent, 182) a protein synthesis inhibitor, 183) an α-glucosidase inhibitor, 184) a calcium channel blocker, 185) a pyruvate dehydrogenase activator, 186) a prostaglandin inhibitor, 187) a sodium channel inhibitor, 188) a serine protease inhibitor, 189) an intracellular calcium flux inhibitor, 190) a JAK2 inhibitor; 191) an androgen inhibitor, 192) an aromatase inhibitor, 193) an anti-viral agent, 194) a 5-HT inhibitor, 195) an FXR antagonist, 196) an actin polymerization and stabilization promoter, 197) an AXOR12 agonist, 198) an angiotensin Il receptor agonist, 199) a platelet aggregation inhibitor, 200) a CB1/CB2 receptor agonist, 201) a norepinephrine reuptake inhibitor, 202) a selective serotonin reuptake inhibitor, 203) a reducing agent, 204) Isotretinoin, 205) radicicol, 206) clobetasol propionate, 207) homoharringtonine, 208) trichostatin A, 209) brefeldin A, 210) thapsigargin, 211) dolastatin 15, 212) cerivastatin, 213) jasplakinolide, 214) herbimycin A, 215) pirfenidone, 216) vinorelbine, 217) 17-DMAG, 218) tacrolimus, 219) loteprednol etabonate, 220) juglone, 221) prednisolone, 222) puromycin, 223) 3-BAABE, 224) cladribine, 225) mannose-6-phosphate, 226) 5-azacytidine, 227) Ly333531 (ruboxistaurin), 228) simvastatin, and 229) an immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof. These and other agents are described in more detail herein.
As used herein and in the appended claims, the singular forms "a," "and," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an agent" includes a plurality of such agents, and reference to "the cell" includes reference to one or more cells and equivalents thereof known to those skilled in the art, and so forth. The term "comprising" (and related terms such as "comprise" or "comprises" or "having" or "including" and the like) is not intended to exclude that in other certain embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, described herein may "consist of or "consist essentially of the described features.
These and other aspects of the present invention will become evident upon reference to the following detailed description and attached drawings. In addition, various references are set forth herein which describe in more detail certain procedures and/or compositions (e.g., polymers), and are therefore incorporated by reference in their entirety.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1A schematically depicts the transcriptional regulation of matrix metalloproteinases.
Figure 1B is a blot which demonstrates that IL-1 stimulates AP-1 transcriptional activity.
Figure 1C is a graph which shows that IL-1 induced binding activity decreased in lysates from chondrocytes which were pretreated with paclitaxel.
Figure 1 D is a blot which shows that IL-1 induction increases collagenase and stromelysin in RNA levels in chondrocytes, and that this induction can be inhibited by pretreatment with paclitaxel.
Figures 2A-H are blots that show the effect of various anti- microtubule agents in inhibiting collagenase expression.
Figure 3 is a graph showing the results of a screening assay for assessing the effect of paclitaxel on smooth muscle cell migration.
Figure 4 is a bar graph showing the area of granulation tissue in carotid arteries exposed to silk coated perivascular polyurethane (PU) films relative to arteries exposed to uncoated PU films.
Figure 5 is a bar graph showing the area of granulation tissue in carotid arteries exposed to silk suture coated perivascular PU films relative to arteries exposed to uncoated PU films.
Figure 6 is a bar graph showing the area of granulation tissue in carotid arteries exposed to natural and purified silk powder and wrapped with perivascular PU film relative to a control group in which arteries are wrapped with perivascular PU film only.
Figure 7 is a bar graph showing the area of granulation tissue (at 1 month and 3 months) in carotid arteries sprinkled with talcum powder and wrapped with perivascular PU film relative to a control group in which arteries are wrapped with perivascular PU film only.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
Prior to setting forth the invention, it may be helpful to an understanding thereof to first set forth definitions of certain terms that are used hereinafter.
"Medical device", "implant", "device", "medical device," "medical implant", "implant/device", and the like are used synonymously to refer to any object that is designed to be placed partially or wholly within a patient's body for one or more therapeutic or prophylactic purposes such as for restoring physiological function, alleviating symptoms associated with disease, delivering therapeutic agents, detecting changes (or levels) in the internal environment, and/or repairing or replacing or augmenting etc. damaged or diseased organs and tissues. While medical devices are normally composed of biologically compatible synthetic materials (e.g., medical-grade stainless steel, titanium and other metals; exogenous polymers, such as polyurethane, silicon, PLA, PLGA), other materials may also be used in the construction of the medical device or implant. Specific medical devices and implants that are particularly useful for the practice of this invention include devices and implants designed to deliver therapeutic levels of a drug to a target tissue (drug delivery pumps) and/or sensors designed to detect changes in body function and/or levels of key physiological metabolites, chemistry, hormones or biological factors.
"Implantable sensor" refers to a medical device that is implanted in the body to detect blood or tissue levels of a particular chemical (e.g., glucose, electrolytes, drugs, hormones) and/or changes in body chemistry, metabolites, function, pressure, flow, physical structure, electrical activity or other variable parameter. Implantable sensors may have one or more electrodes that extend into the external environment to sense a variety of physical and/or physiological properties, including, but not limited to, optical, mechanical, baro, chemical and electrochemical properties. Sensors may be used to detect information, for example, about temperature, strain, pressure, magnetic, acceleration, ionizing radiation, acoustic wave or chemical changes (e.g., blood constituents, such as glucose). For example for the detection of glucose levels, the sensor may utilize an enzyme-based electrochemical sensor, a glucose-responsive hydrogel combined with a pressure sensor, microwires with electrodes, radiofrequency microelectronics and a glucose affinity polymer combined with physical and biochemical sensor technology, and near or mid infrared light emission combined with optical spectroscopy detectors to name a few. Representative examples of implantable sensors include, blood/tissue glucose monitors, electrolyte sensors, blood constituent sensors, temperature sensors, pH sensors, optical sensors, amperometric sensors, pressure sensors, biosensors, sensing transponders, strain sensors, activity sensors and magnetoresistive sensors. "Drug-delivery pump" refers to a medical device that includes a pump which is configured to deliver a biologically active agent (e.g., a drug) at a regulated dose. These devices are implanted within the body and may include an external transmitter for programming the controlled release of drug, or alternatively, may include an implantable sensor that provides the trigger for the drug delivery pump to release drug as physiologically required. Drug-delivery pumps may be used to deliver virtually any agent, but specific examples include insulin for the treatment of diabetes, medication for the relief of pain, chemotherapy for the treatment of cancer, anti-spastic agents for the treatment of movement and muscular disorders, or antibiotics for the treatment of infections. Representative examples of drug delivery pumps for use in the practice of the invention include, without limitation, constant flow drug delivery pumps, programmable drug delivery pumps, intrathecal pumps, implantable insulin delivery pumps, implantable osmotic pumps, ocular drug delivery pumps and implants, metering systems, peristaltic (roller) pumps, electronically driven pumps, elastomeric pumps, spring-contraction pumps, gas-driven pumps (e.g., induced by electrolytic cell or chemical reaction), hydraulic pumps, piston- dependent pumps and non-piston-dependent pumps, dispensing chambers, infusion pumps, passive pumps, infusate pumps and osmotically-driven fluid dispensers.
"Fibrosis," "scarring," or "fibrotic response" refers to the formation of fibrous (scar) tissue in response to injury or medical intervention.
Therapeutic agents which inhibit fibrosis or scarring can do so through one or more mechanisms including: inhibiting the inflammatory response, inhibiting migration or proliferation of connective tissue cells (such as fibroblasts, smooth muscle cells, and vascular smooth muscle cells), inhibiting angiogenesis, reducing ECM production (or promoting ECM breakdown), and/or inhibiting tissue remodeling. In addition, numerous therapeutic agents described in this invention will have the additional benefit of also reducing tissue regeneration (the replacement of injured cells by cells of the same type) when appropriate. "Inhibit fibrosis", "reduce fibrosis", "fibrosis-inhibitor", "inhibits scar", "reduces scar", "anti-fibrosis", "anti-fibrotic," "anti-scarring" and the like are used synonymously to refer to the action of agents or compositions which result in a statistically significant decrease in the formation of fibrous tissue that may be expected to occur in the absence of the agent or composition.
"Inhibitor" refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process. The process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
"Antagonist" refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process. While the process may be a general one, typically this refers to a drug mechanism where the drug competes with a molecule for an active molecular site or prevents a molecule from interacting with the molecular site. In these situations, the effect is that the molecular process is inhibited.
"Agonist" refers to an agent which stimulates a biological process or rate or degree of occurrence of a biological process. The process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
"Anti-microtubule agents" may be understood to include any protein, peptide, chemical, or other molecule which impairs the function of microtubules, for example, through the prevention or stabilization of polymerization. Compounds that stabilize polymerization of microtubules are referred to herein as "microtubule stabilizing agents." A wide variety of methods may be utilized to determine the anti-microtubule activity of a particular compound, including for example, assays described by Smith et al. (Cancer Lett 79(2):213-219, 1994) and Mooberry et al., {Cancer Lett. 96(2):261- 266, 1995).
"Host", "person", "subject", "patient" and the like are used synonymously to refer to the living being (human or animal) into which a device of the present invention is implanted. "Implanted" refers to having completely or partially placed a device within a host. A device is partially implanted when some of the device reaches, or extends to the outside of, a host.
"Release of an agent" refers to a statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the implant/device.
"Biodegradable" refers to materials for which the degradation process is at least partially mediated by, and/or performed in, a biological system. "Degradation" refers to a chain scission process by which a polymer chain is cleaved into oligomers and monomers. Chain scission may occur through various mechanisms, including, for example, by chemical reaction (e.g., hydrolysis) or by a thermal or photolytic process. Polymer degradation may be characterized, for example, using gel permeation chromatography (GPC), which monitors the polymer molecular mass changes during erosion and drug release. Biodegradable also refers to materials may be degraded by an erosion process mediated by, and/or performed in, a biological system. "Erosion" refers to a process in which material is lost from the bulk. In the case of a polymeric system, the material may be a monomer, an oligomer, a part of a polymer backbone, or a part of the polymer bulk. Erosion includes (i) surface erosion, in which erosion affects only the surface and not the inner parts of a matrix; and (ii) bulk erosion, in which the entire system is rapidly hydrated and polymer chains are cleaved throughout the matrix. Depending on the type of polymer, erosion generally occurs by one of three basic mechanisms (see, e.g., Heller, J., CRC Critical Review in Therapeutic Drug Carrier Systems (1984), 1(1), 39- 90); Siepmann, J. et al., Adv. Drug Del. Rev. (2001), 48, 229-247): (1) water- soluble polymers that have been insolubilized by covalent cross-links and that solubilize as the cross-links or the backbone undergo a hydrolytic cleavage; (2) polymers that are initially water insoluble are solubilized by hydrolysis, ionization, or pronation of a pendant group; and (3) hydrophobic polymers are converted to small water-soluble molecules by backbone cleavage. Techniques for characterizing erosion include thermal analysis (e.g., DSC), X- ray diffraction, scanning electron microscopy (SEM), electron paramagnetic resonance spectroscopy (EPR), NMR imaging, and recording mass loss during an erosion experiment. For microspheres, photon correlation spectroscopy (PCS) and other particles size measurement techniques may be applied to monitor the size evolution of erodible devices versus time. As used herein, "analogue" refers to a chemical compound that is structurally similar to a parent compound, but differs slightly in composition (e.g., one atom or functional group is different, added, or removed). The analogue may or may not have different chemical or physical properties than the original compound and may or may not have improved biological and/or chemical activity. For example, the analogue may be more hydrophilic or it may have altered reactivity as compared to the parent compound. The analogue may mimic the chemical and/or biologically activity of the parent compound (i.e., it may have similar or identical activity), or, in some cases, may have increased or decreased activity. The analogue may be a naturally or non- naturally occurring (e.g., recombinant) variant of the original compound. An example of an analogue is a mutein (i.e., a protein analogue in which at least one amino acid is deleted, added, or substituted with another amino acid). Other types of analogues include isomers (enantiomers, diasteromers, and the like) and other types of chiral variants of a compound, as well as structural isomers. The analogue may be a branched or cyclic variant of a linear compound. For example, a linear compound may have an analogue that is branched or otherwise substituted to impart certain desirable properties (e.g., improve hydrophilicity or bioavailability).
As used herein, "derivative" refers to a chemically or biologically modified version of a chemical compound that is structurally similar to a parent compound and (actually or theoretically) derivable from that parent compound. A "derivative" differs from an "analogue" in that a parent compound may be the starting material to generate a "derivative," whereas the parent compound may not necessarily be used as the starting material to generate an "analogue." A derivative may or may not have different chemical or physical properties of the parent compound. For example, the derivative may be more hydrophilic or it may have altered reactivity as compared to the parent compound. Derivatization (i.e., modification) may involve substitution of one or more moieties within the molecule (e.g., a change in functional group). For example, a hydrogen may be substituted with a halogen, such as fluorine or chlorine, or a hydroxyl group (-OH) may be replaced with a carboxylic acid moiety (-COOH). The term "derivative" also includes conjugates, and prodrugs of a parent compound (i.e., chemically modified derivatives which can be converted into the original compound under physiological conditions). For example, the prodrug may be an inactive form of an active agent. Under physiological conditions, the prodrug may be converted into the active form of the compound. Prodrugs may be formed, for example, by replacing one or two hydrogen atoms on nitrogen atoms by an acyl group (acyl prodrugs) or a carbamate group (carbamate prodrugs). More detailed information relating to prodrugs is found, for example, in Fleisher et al., Advanced Drug Delivery Reviews 19 (1996) 115; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; or H. Bundgaard, Drugs of the Future 16 (1991) 443. The term "derivative" is also used to describe all solvates, for example hydrates or adducts (e.g., adducts with alcohols), active metabolites, and salts of the parent compound. The type of salt that may be prepared depends on the nature of the moieties within the compound. For example, acidic groups, for example carboxylic acid groups, can form, for example, alkali metal salts or alkaline earth metal salts (e.g., sodium salts, potassium salts, magnesium salts and calcium salts, and also salts with physiologically tolerable quaternary ammonium ions and acid addition salts with ammonia and physiologically tolerable organic amines such as, for example, triethylamine, ethanolamine or tris-(2-hydroxyethyl)amine). Basic groups can form acid addition salts, for example with inorganic acids such as hydrochloric acid, sulfuric acid or phosphoric acid, or with organic carboxylic acids and sulfonic acids such as acetic acid, citric acid, benzoic acid, maleic acid, fumaric acid, tartaric acid, methanesulfonic acid or p-toluenesulfonic acid. Compounds which simultaneously contain a basic group and an acidic group, for example a carboxyl group in addition to basic nitrogen atoms, can be present as zwitterions. Salts can be obtained by customary methods known to those skilled in the art, for example by combining a compound with an inorganic or organic acid or base in a solvent or diluent, or from other salts by cation exchange or anion exchange. Any concentration ranges, percentage range, or ratio range recited herein are to be understood to include concentrations, percentages or ratios of any integer within that range and fractions thereof, such as one tenth and one hundredth of an integer, unless otherwise indicated. Also, any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness, are to be understood to include any integer within the recited range, unless otherwise indicated. It should be understood that the terms "a" and "an" as used above and elsewhere herein refer to "one or more" of the enumerated components. For example, "a" polymer refers to one polymer or a mixture comprising two or more polymers. As used herein, the term "about" means ± 15%. As discussed above, the present invention provides compositions, methods and devices relating to medical devices and implants (specifically implantable pumps and sensors), which greatly increase their ability to inhibit the formation of reactive scar tissue on, or around, the surface of the device or implant. Described in more detail below are methods for constructing medical devices or implants, compositions and methods for generating medical devices and implants which inhibit fibrosis, and methods for utilizing such medical devices and implants.
A. Clinical Applications of Implantable Sensor and Pump Devices Which Include and Release a Fibrosis-lnhibitinq Agent
1. Implantable Sensors
In one aspect, implantable sensors that include an anti-scarring agent are provided that can be used to detect physiological levels or changes in the body. There are numerous sensor devices where the occurrence of a fibrotic reaction will adversely affect the functioning of the device or the biological problem for which the device was implanted or used. Proper clinical functioning of an implanted sensor is dependent upon intimate anatomical contact with the target tissues and/or body fluids. Scarring around the implanted device may degrade the electrical components and characteristics of the device-tissue interface, and the device may fail to function properly. The formation of scar tissue between the sensing device and the adjacent (target) tissue can prevent the flow of physical, chemical and/or biological information (e.g., fluid levels, drug levels, metabolite levels, glucose levels, pressure etc.) from reaching the detection mechanism of the sensor. Similarly if a "foreign body" response occurs and causes the implanted sensor to become encapsulated by scar (i.e., the body "walls off' the sensor with fibrous tissue), the sensor will receive biological information that is not reflective of the organism as a whole. If the sensor is detecting conditions inside the capsule (i.e., levels detected in a microenvironment), and these conditions are not consistent with those outside the capsule (i.e., within the body as a whole - the microenvironment), it will record information that is not representative of systemic levels.
Sensors or transducers may be located deep within the body for monitoring a variety of physiological properties, such as temperature, pressure, strain, fluid flow, metabolite levels (e.g., electrolytes, glucose), drug levels, chemical properties, electrical properties, magnetic properties, and the like. Representative examples of implantable sensors for use in the practice of the invention include, blood and tissue glucose monitors, electrolyte sensors, blood constituent sensors, temperature sensors, pH sensors, optical sensors, amperometric sensors, pressure sensors, biosensors, sensing transponders, strain sensors, activity sensors and magnetoresistive sensors.
Numerous types of implantable sensors and transducers have been described. For example, the implantable sensor may be a microelectronic device that is implanted around the large bowels to control bowel function by detecting rectal contents and stimulating peristaltic contractions to empty the bowels when it is convenient. See, e.g., U.S. Patent No. 6,658,297. The implantable sensor may be used to measure pH in the Gl tract. A representative example of such a pH sensing device is the BRAVO pH Monitoring System from Medtronic, Inc. (Minneapolis, MN). The implantable sensor may be part of a Gl catheter or probe that includes a sensor portion connected to an electrical or optical measurement device and a sensitive polymeric material that undergoes an irreversible change when exposed to cumulative action of an external medium. See, e.g., U.S. Patent No. 6,006,121. The implantable sensor may be a component of a central venous catheter (CVC) (e.g., a jugular vein catheter) system. For example, the device may be composed of a catheter body having at least one oxygen sensor and a distal heat exchange region in which the catheter body is formed with coolant supply and return lumens to provide heat exchange within a body to prevent overheating due to severe brain trauma or ischemia due to stroke. See, e.g., U.S. Patent No. 6,652,565. A CVC may include a thermal mass and a temperature sensor to measure blood temperature. See, e.g., U.S. Patent No. 6,383,144.
Several specific implantable sensor devices and treatments will be described in greater detail including:
a. Blood and Glucose Monitors Glucose monitors are used to detect changes in blood glucose, specifically for the management and treatment of patients with diabetes mellitus. Diabetes is a metabolic disorder of glucose metabolism that afflicts tens of millions of people in the developed countries of the world. This disease is characterized by the inability of the body to properly utilize and metabolize carbohydrates, particularly glucose. Normally, the finely-tuned balance between glucose in the blood and glucose in the bodily tissue cells is maintained by insulin, a hormone produced by the pancreas. If the pancreas becomes defective and insulin is produced in inadequate amounts to reduce blood glucose levels (Type I diabetes), or if the body becomes insensitive to the glucose-lowering effects of insulin despite adequate pancreatic insulin production (Type Il diabetes), the result is diabetes. Accurate detection of blood glucose levels is essential to the management of diabetic patients because the dosage and timing of administration of insulin and/or other hypoglycemic agents are titrated depending upon changes in glucose levels in response to the medication. If the dosage is too high, blood glucose levels drop too low, resulting in confusion and potentially even loss of consciousness. If the dosage is too low, blood glucose levels rise too high, leading to excessive thirst, urination, and changes in metabolism known as ketoacidosis. If the timing of medication administration is incorrect, blood glucose levels can fluctuate wildly between the two extremes - a situation that is thought to contribute to some of the long-term complications of diabetes such as heart disease, kidney failure and blindness. Since in the extreme, all these conditions can be life threatening, careful and continuous monitoring of glucose levels is a critical aspect of diabetes management. One way to detect changes in glucose levels and to continuously sense when levels of glucose become too high or too low in diabetes patients is to implant a glucose sensor. As the glucose sensor detects changes in the blood glucose levels, insulin can be administered by external injection or via an implantable insulin pump to maintain blood glucose levels within an acceptable physiologic range. Numerous types of blood and tissue glucose monitors are suitable for use in the practice of the invention. For example, the glucose monitor may be delivered to the vascular system transluminal^ using a catheter on a stent platform. See, e.g., U.S. Patent No. 6,442,413. The glucose monitor may be composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations. See, e.g., U.S. Patent Nos. 5,101 ,814 and 5,190,041. The glucose monitor may be a small diameter flexible electrode implanted subcutaneously which may be composed of an analyte-responsive enzyme designed to be an electrochemical glucose sensor. See, e.g., U.S. Patent Nos. 6,121 ,009 and 6,514,718. The implantable sensor may be a closed loop insulin delivery system whereby there is a sensing means that detects the patient's blood glucose level based on electrical signals and then stimulates either an insulin pump or the pancreas to supply insulin. See, e.g., U.S. Patent Nos. 6,558,345 and 6,093,167. Other glucose monitors are described in, for e.g., U.S. Patent Nos. 6,579,498; 6,565,509 and 5,165,407. Minimally invasive glucose monitors include the GLUCOWATCH G2 BIOGRAPHER from Cygnus Inc. (see cygn.com); see, e.g., U.S. Patent Nos. 6,546,269; 6,687,522; 6,595,919 and U.S. Patent Application Nos. 20040062759A1 ; 20030195403A1; and 20020091312A1. Numerous commercially available blood and tissue glucose sensor devices are suitable for the practice of this invention. Although virtually any implantable glucose sensor may be utilized, several specific commercial and development stage examples are described below for greater clarity.
The CONTINUOUS GLUCOSE MONITORING SYSTEM (CGMS) from Medtronic MiniMed, Inc. (Northridge, CA; see minimed.com); see, e.g., U.S. Patent Nos. 6,520,326; 6,424,847; 6,360,888; 5,605,152; 6,804,544; and U.S. Patent Application No. 20040167464A1. The CGMS system is surgically implanted in the subcutaneous tissue of the abdomen and stores tissue glucose readings every 5 minutes. Coating the sensor with a fibrosis-inhibiting agent may prolong the activity of this device because it often must be removed after several days (approximately 3), in part because it loses its sensitivity as a result of the local tissue reaction to the device.
The CONTINUOUS GLUCOSE MONITORING DEVICE from TheraSense (Alameda, CA, see therasense.com) which utilizes a disposable, miniaturized electrochemical sensor that is inserted under the patient's skin using a spring-loaded insertion device. The sensor measures glucose levels in the interstitial fluid every five minutes, with the ability to store results for future analysis. See, e.g., US20040186365A1 ; US20040106858A1 and US20030176183A1. Even though the device can store up to a month of data and has alarms for high and low glucose levels, it must be replaced every few days because it loses its accuracy as a result of the foreign body reaction to the implant. Utilizing this sensor in combination with a fibrosis-inhibiting agent may prolong its activity, enhance its performance and reduce the frequency of replacement. Another electrochemical sensor that may benefit from the present invention is the multilayered implantable electrochemical sensor from lsense (Portland, OR). This system consists of a semipermeable membrane, a catalytic membrane which generates an electrical current in the presence of glucose, and a specificity membrane to reduce interference from other substances.
The SMSI glucose sensor (Sensors for Medicine and Sciences, Inc., Montgomery County, Maryland; see s4ms.com) is designed to be implanted under the skin in a short outpatient procedure. The sensor is designed to automatically measure interstitial glucose every few minutes, without any user intervention. The sensor implant communicates wirelessly with a small external reader, allowing the user to monitor glucose levels continuously or on demand. The reader is designed to be able to track the rate of change of glucose levels and warn the user of impending hypo- or hyperglycemia. The operational life of the sensor implant is about 6-12 months, after which it may be replaced.
Animas Corporation (West Chester, PA; animascorp.com) is developing an implantable glucose sensor that measures the near-infrared absorption of blood based on spectroscopy or optical sensing placed around a vein. The Animas glucose monitor may be tied to an insulin infusion pump to provide a closed-loop control of blood glucose levels. Scar tissue over the sensor distorts the ability of the device to correctly gather optical information and may thus benefit from use in combination with a fibrosis inhibiting agent. DexCom, Inc. (San Diego, CA; see dexcom.com) is developing their Continuous Glucose Monitoring System which is an implantable sensor that wirelessly transmits continuous blood glucose readings to an external receiver. The receiver displays the current glucose value every 30 seconds, as well as one-hour, three-hour and nine-hours trended values, and sounds an alert when a high or low glucose excursion is detected. This device features an implantable sensor that is placed in the subcutaneous tissue and continuously monitors tissue (interstitial fluid) glucose levels for both type 1 and type 2 diabetics. This device may also include a unique microarchitectural arrangement in the sensor region that allows accurate data to be obtained over long periods of time. Glucose monitoring devices and associated systems that are developed by DexCom, Inc. are described in, for example, U.S. Patent Nos. 6,741 ,877; 6,702,857 and 6,558,321. Unfortunately, even though the battery and circuitry of monitoring devices allows long-term functioning, a foreign body response and/or encapsulation of the implant affect the ability of the device to detect glucose levels accurately for prolonged periods in a percentage of implants. Combining this device with an inhibitor of fibrosis (e.g., by coating the implant and/or sensor with the agent, incorporating the agent into the polymers that make up the implant, and/or infiltrating it into the tissue surrounding the implant) may allow it to accurately detect glucose levels for longer periods of time after implantation, reduce the number of devices that fail and decrease the incidence of replacement.
Also of particular interest in the practice of this invention is glucose monitoring systems that utilize a glucose-responsive polymer as part of their detection mechanism. M-Biotech (Salt Lake City, UT) is developing a continuous monitoring system that consists of subcutaneous implantation of a glucose-responsive hydrogel combined with a pressure transducer. See, e.g., U.S. Patent Nos.; and. The hydrogel responds to changes in glucose concentration by either shrinking or swelling and the expansion or contraction is detected by the pressure transducer. The transducer converts the information into an electrical signal and sends a wireless signal to a display device. Cybersensors (Berkshire, UK) produces a capsule-like sensor implanted under the skin and an external receiver/transmitter that captures the data and powers the capsule via RF signals (see, e.g., GB 2335496 and U.S. Patent No. 6,579,498) Issued by the UK Patent and Trademark Office). The sensor capsule is composed of a glucose affinity polymer and contains a physical sensor and an RF microchip; the entire capsule is further enclosed in a semipermeable membrane. The glucose affinity polymer exhibits rheological changes when exposed to glucose (in the range of 3-15 nM) by becoming thinner and less viscous as glucose concentrations increase. This reversible reaction can be detected by the physical sensor and converted into a signal. These aforementioned systems offer an excellent opportunity for combining the implanted sensor with fibrosis-inhibiting agents and compositions. Not only can the agent be coated onto the surface of the sensor or infiltrated into the tissue surrounding the sensor, but it can also be incorporated into the glucose- responsive hydrogels and polymers that make up the implant. Another glucose sensing device is under development by
Advanced Biosensors (Mentor, OH) that consists of small (150 μm wide by 2 mm long), biocompatible, silicon-based needles that are implanted under the skin. The device senses glucose levels in the dermis and transmits data wirelessly. Unfortunately, a foreign body response and/or encapsulation of the implant affect the ability of the device to detect glucose levels accurately for longer than 7 days. Combining this device with an inhibitor of fibrosis may allow it to accurately detect glucose levels for longer periods of time and extend the effective lifespan of the device.
Regardless of the specific design features of implantable blood, tissue, or interstitial fluid glucose sensor devices, for accurate detection of physical, chemical and/or physiological properties, the device must be accurately positioned adjacent to the tissue. In particular, the detector of the sensing mechanism must be exposed to glucose levels that are identical to (or representative of) those found in the bloodstream. If excessive scar tissue growth or extracellular matrix deposition occurs around the device, this can impair the movement of glucose from the tissue to the detector and render it ineffective. Similarly if a "foreign body" response occurs and causes the implanted glucose sensor to become encapsulated by fibrous tissue, the sensor will be detecting glucose levels in the capsule. If glucose levels inside the capsule are not consistent with those outside the capsule (i.e., within the body as a whole), it will record information that is not representative of systemic levels. This can cause the physician or the patient to administer the wrong dosage of hypoglycemic drugs (such as insulin) with potentially serious consequences. Blood, tissue or interstitial fluid glucose sensor devices that release a therapeutic agent able to reduce scarring and/or encapsulation of the implant can increase the efficiency and accuracy of glucose detection, minimize insulin dosing errors, assist in the maintenance of correct blood glucose levels, increase the duration that these devices function clinically, and/or reduce the frequency of implant replacement. In one aspect, the device includes blood, tissue and interstitial fluid glucose monitoring devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent. The fibrosis-inhibiting agent can also be incorporated into, and released from, the components of the implanted sensor. This embodiment is particularly useful for implants employing glucose-responsive polymers and hydrogels (that can be drug-loaded with an active agent) as well as those utilizing a semi-permeable membrane around the sensor (which can also be loaded with a fibrosis- inhibiting agent). As an alternative to this, or in addition to this, a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the glucose sensor is, or will be, implanted.
b. Pressure and Stress Sensors
In another aspect, the implantable sensor may be a pressure monitor. Pressure monitors may be used to detect increasing pressure or stress within the body. Implantable pressure transducers and sensors are used for temporary or chronic use in a body organ, tissue or vessel for recording absolute pressure. Many different designs and operating systems have been proposed and placed into temporary or chronic use for patients with a variety of medical conditions. Indwelling pressure sensors for temporary use of a few days or weeks are available, however, chronically or permanently implantable pressure sensors have also been used. Pressure sensors may detect many types of bodily pressures, such as, but not limited to blood pressure and fluid flow, pressure within aneurysm sacs, intracranial pressure, and mechanical pressure associated with bone fractures.
Numerous types of pressure monitors are suitable for use in the practice of the invention. For example, the implantable sensor may detect body fluid absolute pressure at a selected site and ambient operating temperature by using a lead, sensor module, sensor circuit (including electrical conductors) and means for providing voltage. See, e.g., U.S. Patent No. 5,535,752. The implantable sensor may be an intracranial pressure monitor that provides an analogue data signal which is converted electronically to a digital pulse. See, e.g., U.S. Patent No. 6,533,733. The implantable sensor may be a barometric pressure sensor enclosed in an air chamber which is used for deriving reference pressure data for use in combination with an implantable medical device, such as a pacemaker. See, e.g., U.S. Patent No. 6,152,885. The implantable sensor may be adapted to be inserted into a body passageway to monitor a parameter related to fluid flow through an endoluminal implant (e.g., stent). See, e.g., U.S. Patent No. 5,967,986. The implantable sensor may be a passive sensor with an inductor-capacitor circuit having a resonant frequency which is adapted for the skull of a patient to sense intracranial pressure. See, e.g., U.S. Patent No. 6,113,553. The implantable sensor may be a self- powered strain sensing system that generates a strain signal in response to stresses that may be produced at a bone fixation device. See, e.g., U.S. Patent No. 6,034,296. The implantable sensor may be a component of a perfusion catheter. The catheter may include a wire electrode and a lumen for perfusing saline around the wire, which is designed for measuring a potential difference across the Gl wall and for simultaneous measurement of pressure. See, e.g., U.S. Patent No. 5,551 ,425. The implantable sensor may be part of a CNS device; for example, an intracranial pressure sensor which is mounted within the skull of a body at the situs where the pressure is to be monitored and a means of transmitting the pressure externally from the skull. See, e.g., U.S. Patent No. 4,003,141. The implantable sensor may be a component of a left ventricular assist device. For example, the VAD may be a blood pump adapted to be joined in flow communication between the left ventricle and the aorta using an inlet flow pressure sensor and a controller that may adjust speed of pump based on sensor feedback. See, e.g., U.S. Patent No. 6,623,420. Numerous commercially available and experimental pressure and stress sensor devices are suitable for the practice of the invention. By way of illustration, a selection of these devices and implants are described in the following paragraphs
A device from CardioMEMS (Atlanta, GA; @cardiomems.com, a partnership between the Georgia Institute of Technology and the Cleveland Clinic) which can be inserted into an aneurysm sac to monitor pressure within the sac and thereby alert a medical specialist to the filing of the sac with fluid, possibly to rupture-provoking levels. Endovascular aneurysm repair (EVAR) is often performed using a stent graft which isolates the aneurysm from the circulation. However, persistent leakage of blood into the aneurysm sac results in ongoing pressure build-up in the sac and a resultant risk of rupture. The CardioMEMS device is implanted into the aneurysm sac after EVAR to monitor pressure in the isolated sac in order to detect which patients are at increasing risk of rupture. The pressure sensor features an inductive-capacitive resonant circuit with a variable capacitor. Since capacitance varies with the pressure in the environment in which the capacitor is placed, it can detect changes in local pressure. Data is generated by using external excitation systems that induce an oscillating current in the sensor and detecting the frequency of oscillation (which is then used to calculate pressure). Unfortunately, even though the circuitry allows long-term functioning, a foreign body response and/or encapsulation of the implant affect the ability of the device to detect accurate pressure levels in the aneurysm (i.e., the device detects the pressure in the microenvironment of the capsule, not of the aneurysm sac as a whole).
Combining this device with an inhibitor of fibrosis (e.g., by coating the implant and/or sensor with the agent, incorporating the agent into the polymers that make up the implant, and/or infiltrating it into the sac surrounding the implant) may allow it to accurately detect pressure levels for longer periods of time after implantation and reduce the number of devices that fail.
MicroStrain Inc. (Williston, VT, @microstrain.com) has developed a family of wireless implantable sensors for measuring strain, position and motion within the body. These sensors can measure, for example, eye tremor, depth of corneal implant, orientation sensor for improved tooth crown prep, mayer ligament strains, spinal ligament strains, vertebral bone strains, elbow ligament strains, emg and ekg data, 3DM-G for measurement of orientation and motion, wrist ligament strains, hip replacement sensors for measuring micromotion, implant subsidence, knee ligament strain, ankle ligament strain, Achilles tendon strain, foot arch support strains, force within foot insoles. The company provides a knee prosthesis that can measure in vivo compressive forces and transmit the data in real time. Patents describing this technology, and components used in the manufacture of devices for this technology include US 6,714,763; 6,625,517; 6,622,567; 6,588,282; 6,529,127; 6,499,368; 6,433,629; 5,887,351 ; 5,777,467; 5,497,147; and 4,993,428. US Patent Applications describing this technology, and components used in the manufacture of devices for this technology include 20040113790;
20040078662; 20030204361 ; 20030158699; 20030047002; 20020190785; 20020170193; 20020088110; 20020085174; 20010054317; and 20010033187. Mesotec (Hannover, Germany; @mesotec.com), in collaboration with several German institutes (e.g., Fraunhofer Institute of Microelectronic Circuits and Systems), has developed an implantable intraocular pressure sensor system, called the MESOGRAPH, which can continuously monitor intraocular pressure. This is desirable, e.g., in order to identify the onset of glaucoma. The CMOS-based sensor can be implanted during standard surgical procedures and is inductively linked to an external unit integrated into a spectacle frame. The glasses are in turn linked via a cable to a portable data logger. Data is relayed upstream to the glasses using a modulated RF carrier operating at 13.56 MHz and a switchable load, while power comes downstream to the sensor. By varying the diameter of the polysilicon diaphragms in the on- chip micromechanical vacuum gap capacitors, the pressure range to which the sensor responds can be adapted between 50kNm-2 and 3.5MNm-2. The device consists of a fine, foldable coil for telemetric coupling and a very small miniaturized pressure sensor. The sensor is manufactured on a micro- technological basis and serves for continuous, long-term reading and monitoring of intraocular pressure. Chip and coil are integrated in modified soft intraocular lenses, which can be implanted in the patient's eye during today's common surgical procedures. Unfortunately, the device often fails after initially successful implantation because a foreign body response and/or encapsulation of the implant affect the ability of it to detect accurate pressure levels in the eye (Ae., the device detects the pressure in the microenvironment of the capsule surrounding the implant, not intraocular pressure as a whole). Combining this device with an inhibitor of fibrosis (e.g., by coating the implant and/or sensor with the agent, incorporating the agent into the polymers that make up the implant, and/or infiltrating it into the eye tissue surrounding the implant) may allow it to accurately detect pressure levels for longer periods of time after implantation and reduce the number of devices that fail. Regardless of the specific design features of the pressure or stress sensor, for accurate detection of physical and/or physiological properties (such as pressure), the device must be accurately positioned within the tissue and receive information that is representative of conditions as a whole. If excessive scar tissue growth or extracellular matrix deposition occurs around the device, the sensor may receive erroneous information that compromises its efficacy or the scar tissue may block the flow of biological information to the sensor. For example, many devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant pressure levels (i.e., the device detects the pressure in the microenvironment of the capsule surrounding the implant, not the pressure of the larger environment). Pressure and stress sensing devices that release a therapeutic agent able to reduce scarring can increase the efficiency of detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent. The fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted sensor. As an alternative to this, or in addition to this, a composition that includes an anti- scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
c. Cardiac Sensors
In another aspect, the implantable sensor may be a device configured to detect properties in the heart or in cardiac muscle tissue. Cardiac sensors are used to detect parameters associated with the performance of the heart as monitored at any given time point along a prolonged time period. Typically, monitoring of the heart is often conducted to detect changes associated with heart disease, such as chronic heart failure (CHF). By monitoring patterns associated with heart function, deterioration based on hemodynamic changes can be detected (parameters such as cardiac output, ejection fraction, pressure, ventricular wall motion, etc.). This constant direct monitoring is central to disease management in patients that present with CHF. By monitoring hemodynamic measures directly using implantable sensors, a hemodynamic crisis can be detected and the appropriate medications and interventions selected. Numerous types of cardiac sensors are suitable for use in the practice of the invention. For example, the implantable sensor may be an activity sensor incorporating a magnet and a magnetoresistive sensor that provides a variable activity signal as part of a cardiac device. See, e.g., U.S. Patent No. 6,430,440 and 6,411 ,849. The implantable sensor may monitor blood pressure in a heart chamber by emitting wireless communication to a remote device. See, e.g., U.S. Patent No. 6,409,674. The implantable sensor may be an accelerometer-based cardiac wall motion sensor which transduces accelerations of cardiac tissue to a cardiac stimulation device by using electrical signals. See, e.g., U.S. Patent No. 5,628,777. The implantable sensor may be implanted in the heart's cavity with an additional sensor implanted in a blood vessel to detect pressure and flow within heart's cavity. See, e.g., U.S. Patent No. 6,277,078.
Commercially available cardiac sensor devices suitable for the practice of the invention include Biotronik's (Biotronik GmbH & Co., Berlin, Germany, see biotronik.com) CARDIAC AIRBAG ICD SYSTEM is a rhythm monitoring device that offers rescue shock capability delivering 30 Joule shock therapies for up to 3 episodes of ventricular fibrillation. In addition to the rescue shock capability the system can also provide bradycardia pacing and VT monitoring. The PROTOS family of pacemakers from Biotronik (see biotronikusa.com) also incorporates pacing sensor capability called Closed Loop Simulation. Blood flow and tissue perfusion monitors can be used to monitor noncardϊac tissue as well. Researchers at Oak Ridge National Laboratory have developed a wireless sensor that monitors blood flow to a transplanted organ for the early detection of transplant rejection. Medtronic (Minneapolis, MN; see medtronic.com) is developing their CHRONICLE implantable product, which is designed to continuously monitor a patient's intracardiac pressures, heart rate and physical activity using a sensor placed directly in the heart's chamber. The patient periodically downloads this information to a home-based device that transmits this physiologic data securely over the Internet to a physician.
Regardless of the specific design features of the cardiac sensor, for accurate detection of physical and/or physiological properties (such as pressure, flow rates, etc.), the device must be accurately positioned within the heart muscle, chambers or great vessels and receive information that is representative of conditions as a whole. If excessive scar tissue growth or extracellular matrix deposition occurs around the sensing device, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of biological information to the detector mechanism of the sensor. For example, many cardiac monitoring devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (Ae., the device detects conditions in the microenvironment of the capsule surrounding the implant, not the pressure of the larger environment). Cardiac sensing devices that release a therapeutic agent able to reduce scarring can increase the efficiency of detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices that are coated with an anti- scarring agent or a composition that includes an anti-scarring agent. The fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted cardiac sensor. As an alternative to this, or in addition to this, a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
d. Respiratory Sensors
In another aspect, the implantable sensor may be a device configured to detect properties in the respiratory system. Respiratory sensors may be used to detect changes in breathing patterns. For example, a respiratory sensor may be used to detect sleep apnea, which is an airway disorder. There are two kinds of sleep apnea. In one condition, the body fails to automatically generate the neuromuscular stimulation necessary to initiate and control a respiratory cycle at the proper time. In the other condition, the muscles of the upper airway contract during the time of inspiration and thus the airway becomes obstructed. The cardiovascular consequences of apnea include disorders of cardiac rhythm (bradycardia, auriculoventricular block, ventricular extrasystoles) and hemodynamic disorders (pulmonary and systemic hypertension). This results in a stimulatory metabolic and mechanical effect on the autonomic nervous system and the potential to ultimately lead to increased morbidity. To treat this condition, implantable sensors may be used to monitor respiratory functioning to detect an apnea episode so the appropriate response (e.g., electrical stimulation to the nerves of the upper airway muscles) or other treatment can be provided.
Numerous types of respiratory sensors are suitable for use in the practice of the invention. For example, the implantable sensor may be a respiration element implanted in the thoracic cavity which is capable of generating a respiration signal as part of a ventilation system for providing gas to a host. See, e.g., U.S. Patent No. 6,357,438. The implantable sensor may be composed of a sensing element connected to a lead body which is inserted into bone (e.g., manubrium) that communicates with the intrathoracic cavity to detect respiratory changes. See, e.g., U.S. Patent No. 6,572,543.
Regardless of the specific design features of the respiratory sensor, for accurate detection of physical and/or physiological properties, the device must be accurately positioned adjacent to the tissue. If excessive scar tissue growth or extracellular matrix deposition occurs around the pulmonary function or airway sensing device, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of biological information to the detector mechanism of the sensor. For example, many pulmonary function sensing devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (i.e., the device detects conditions in the microenvironment of the capsule surrounding the implant, not the functioning of the respiratory system as whole). Respiratory sensing devices that release a therapeutic agent able to reduce scarring can increase the efficiency of detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices that are coated with an anti- scarring agent or a composition that includes an anti-scarring agent. The fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted respiratory sensor. As an alternative to this, or in addition to this, a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
e. Auditory Sensors
In another aspect, the implantable sensor may be a device configured to detect properties in the auditory system. Auditory sensors are used as part of implantable hearing systems for rehabilitation of pure sensorineural hearing losses, or combined conduction and inner ear hearing impairments. Hearing systems may include an implantable sensor which delivers an electrical signal which is processed by an implanted processor and delivered to an implantable electromechanical transducer which acts on the middle or inner ear. The auditory sensor acts as the microphone of the hearing system and acts to convert the incident airborne sound into an electrical signal. Numerous types of auditory sensors as part of a hearing system are suitable for use in the practice of the invention. For example, the implantable sensor may generate an electrical audio signal as part of a hearing system for rehabilitation of hearing loss. See, e.g., U.S. Patent No. 6,334,072. The implantable sensor may be a capacitive sensor which is mechanically or magnetically coupled to a vibrating auditory element, such as the malleus, which detects the time-varying capacitance values resulting from the vibrations. See, e.g., U.S. Patent No. 6,190,306. The implantable sensor may be an electromagnetic sensor having a permanent magnet and a coil and a time- varying magnetic flux linkage based on the vibrations which are provided to an output stimulator for mechanical or electrical stimulation of the cochlea. See, e.g., U.S. Patent No. 5,993,376.
Commercially available auditory sensor devices suitable for the practice of the invention include: the HIRES 9OK Bionic Ear Implant, HIRESOLUTION SOUND, CLARION CII Bionic Ear, and CLARION 1.2, from Advanced Bionics (Sylmar, California, a Boston Scientific Company, see advancedbionics.com); see also U.S. Patent Nos. 6,778,858; 6,754,537; 6,735,474; 6,731 ,986; 6,658,302; 6,636,768; 6,631 ,296; 6,628,991 ; 6,498,954; 6,487,453; 6,473,651 ; 6,415,187; and 6,415,185; the NUCLEUS 3 cochlear implant from Cochlear (Lane Cove NSW, Australia, see cochlear.com); see also U.S. Patent Nos. 6,810,289; 6,807,455; 6,788,790; 6,782,619; 6,751,505; 6,736,770; 6,700,982; 6,697,674; 6,678,564; 6,620,093; 6,575,894; 6,570,363; 6,565,503; 6,554,762; 6,537,200; 6,525,512; 6,496,734; 6,480,820; 6,421 ,569; 6,411 ,855; 6,394,947; 6,392,386; 6,377,075; 6,301 ,505; 6,289,246; 6,116,413; 5,720,099; 5,653,742; 5,645,585; and U.S. Patent Application Publication Nos. 2004/0172102A1 and 2002/0138115A1 ; the PULSAR Cl 100 and COMBI 40+ cochlear implants from Med-EI (Austria, see medel.com); see also US Patent Application 20040039245A1 , US Patent Nos. 6,600,955; 6,594,525; 6,556,870; and 5,983,139; the ALLHEAR implants from AIIHear, Inc. (Aurora, Oregon; see allhear.com); see also WO 01/50816; EP 1 245 134; and the DIGISONIC CONVEX, DIGISONIC AUDITORY BRAINSTEM, and DIGISONIC MULTI- ARRAY implants from MXM (France; see mxmlab.com); see also U.S. Patent Nos. 5,123,422; EP 0 219 380; WO 04/002193; EP 1 244 400 A1 ; US 6,428,484; US 20020095194A1 ; WO 01/50992.
Regardless of the specific design features of the auditory sensor, for accurate detection of sound, the device must be accurately positioned within the ear. If excessive scar tissue growth or extracellular matrix deposition occurs around the auditory sensor, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of sound waves to the detector mechanism of the sensor. Auditory sensing devices that release a therapeutic agent able to reduce scarring can increase the efficiency of sound detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent. The fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted auditory sensor. As an alternative to this, or in addition to this, a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
f. Electrolyte and Metabolite Sensors
In another aspect, implantable sensors may be used to detect electrolytes and metabolites in the blood. For example, the implantable sensor may be a device to monitor constituent levels of metabolites or electrolytes in the blood by emitting a source of radiation directed towards blood such that it interacts with a plurality of detectors that provide an output signal. See, e.g., U.S. Patent No. 6,122,536. The implantable sensor may be a biosensing transponder which is composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader. See, e.g., U.S. Patent No. 5,833,603. The implantable sensor may be a monolithic bioelectronic device for detecting at least one analyte within the body of an animal. See, e.g., U.S. Patent No. 6,673,596. Other sensors that measure chemical analytes are described in, e.g., U.S. Patent Nos. 6,625,479 and 6,201 ,980. If excessive scar tissue growth or extracellular matrix deposition occurs around the sensor, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of metabolites or electrolytes to the detector mechanism of the sensor. For example, many metabolite/electrolyte sensing devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (i.e., the device detects conditions in the microenvironment of the capsule surrounding the implant, not blood levels). Sensing devices that release a therapeutic agent able to reduce scarring can increase the efficiency of metabolite/electrolyte detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices that are coated with an anti-scarring agent or a composition that includes an anti-scarring agent. The fibrosis-inhibiting agent can also be incorporated into, and released from, the components (such as polymers) that are part of the structure of the implanted sensor. As an alternative to this, or in addition to this, a composition that includes an anti-scarring agent can be infiltrated into the tissue surrounding where the device is, or will be, implanted.
Although numerous examples of implantable sensor devices have been described above, all possess similar design features and cause similar unwanted foreign body tissue reactions following implantation. It may be obvious to one of skill in the art that commercial sensor devices not specifically cited above as well as next-generation and/or subsequently-developed commercial sensor products are to be anticipated and are suitable for use under the present invention. The sensor device, particularly the sensing element, must be positioned in a very precise manner to ensure that detection is carried out at the correct anatomical location in the body. All, or parts, of a sensor device can migrate following surgery, or excessive scar tissue growth can occur around the implant, which can lead to a reduction in the performance of these devices. The formation of a fibrous capsule around the sensor can impede the flow of biological information to the detector and/or cause the device to detect levels that are not physiologically relevant (i.e., detect levels in the capsule instead of true physiological levels outside the capsule). Not only can this lead to incomplete or inaccurate readings, it can cause the physician or the patient to make incorrect therapeutic decisions based on the information generated. Implantable sensor devices that release a therapeutic agent for reducing scarring (or fibrosis) at the sensor-tissue interface can be used to increase the efficacy and/or the duration of activity of the implant. In one aspect, the present invention provides implantable sensor devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in implantable sensor devices will be described below. These compositions can further include one or more fibrosis-inhibiting agents such that the overgrowth of granulation, fibrous, or neointimal tissue is inhibited or reduced.
Methods for incorporating fibrosis-inhibiting compositions onto or into these sensor devices include: (a) directly affixing to the sensing device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier), (b) directly incorporating into the sensing device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier (c) by coating the sensing device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving a fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the sensing device, (e) by inserting the sensing device into a sleeve or mesh which is comprised of, or coated with, a fibrosis-inhibiting composition, (f) constructing the sensing device itself (or a portion of the device and/or the detector) with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the sensing device surface or to a linker (small molecule or polymer) that is coated or attached to the device (or detector) surface. Each of these methods illustrates an approach for combining the sensor, detector or electrode with a fibrosis-inhibiting (also referred to herein as anti-scarring) agent according to the present invention. For these sensors, detectors and electrodes, the coating process can be performed in such a manner as to: (a) coat a portion of the sensing device (such as the detector); or (b) coat the entire sensing device with the fibrosis-inhibiting composition. In addition to, or alternatively, the fibrosis- inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final product. In these manners, a medical device may be prepared which has a coating, where the coating is, e.g., uniform, non-uniform, continuous, discontinuous, or patterned.
In another aspect, an implantable sensor device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug (Ae., one or more fibrosis-inhibiting agents). The reservoirs may be formed from divets in the device surface or micropores or channels in the device body. In one aspect, the reservoirs are formed from voids in the structure of the device. The reservoirs may house a single type of drug (e.g., fibrosis-inhibiting agent) or more than one type of drug (e.g., a fibrosis-inhibiting agent and an anti-infective agent). The drug(s) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs. The filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier. In certain embodiments, the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount (dose) of drug, and each layer may have a different composition to further tailor the amount and type of drug that is released from the substrate. The multi-layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void. Thus, the coating of the medical device may directly contact the implantable sensor device, or it may indirectly contact the device when there is something, e.g., a polymer layer, that is interposed between the sensor device and the coating that contains the fibrosis-inhibiting agent. In addition to, or as an alternative to, incorporating a fibrosis- inhibiting agent onto or into the implantable sensor device, the fibrosis-inhibiting agent can be applied directly or indirectly to the tissue adjacent to the sensor device (preferably near the sensor-tissue interface). This can be accomplished by applying the fibrosis-inhibiting agent, with or without a polymeric, non- polymeric, or secondary carrier: (a) to the sensor and/or detector surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) prior to, immediately prior to, or during, implantation of the sensor; (c) to the surface of the sensor and/or the tissue surrounding the implanted sensor and/or detector (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the sensor; (d) by topical application of the anti-fibrosis agent into the anatomical space where the implantable sensor will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the implantable sensor as a solution, as an infusate, or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, antiplatelet, and/or anti-infective agents) can also be used.
It may be noted that certain polymeric carriers themselves can help prevent the formation of fibrous tissue on the sensor and/or fibrous encapsulation of the implanted sensor. These carriers (described below) are particularly useful for the practice of this embodiment, either alone, or in combination with a fibrosis-inhibiting composition. The following polymeric carriers can be infiltrated (as described in the previous paragraph) into the vicinity of the sensor-tissue interface and include: (a) sprayable collagen- containing formulations such as COSTASIS and crosslinked derivatized poly(ethylene glycol) -collagen compositions (described, e.g., in U.S. Patent Nos. 5,874,500 and 5,565,519 and referred to herein as "CT3"(both from Angiotech Pharmaceuticals, Inc., Canada), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the detector/sensor surface); (b) sprayable PEG-containing formulations such as COSEAL (Angiotech Pharmaceuticals, Inc.), FOCALSEAL (Genzyme Corporation, Cambridge, MA), SPRAYGEL or DURASEAL (both from Confluent Surgical, Inc., Boston, MA), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the detector/sensor surface); (c) fibrinogen- containing formulations such as FLOSEAL or TISSEAL (both from Baxter Healthcare Corporation, Fremont, CA), either alone, or loaded with a fibrosis- inhibiting agent, applied to the implantation site (or the detector/sensor surface);
(d) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE (both from Q-Med AB, Sweden), HYLAFORM (Inamed Corporation, Santa Barbara, CA), SYNVISC (Biomatrix, Inc., Ridgefield, NJ), SEPRAFILM or SEPRACOAT (both from Genzyme Corporation), loaded with a fibrosis- inhibiting agent applied to the implantation site (or the detector/sensor surface);
(e) polymeric gels for surgical implantation such as REPEL (Life Medical Sciences, Inc., Princeton, NJ) or FLOWGEL (Baxter Healthcare Corporation) alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the detector/sensor surface); (f) orthopedic "cements" used to hold prostheses and tissues in place loaded with a fibrosis-inhibiting agent applied to the implantation site (or the detector/sensor surface), such as OSTEOBOND (Zimmer, Inc., Warsaw, IN), low viscosity cement (LVC) from Wright Medical Technology, Inc. (Arlington, TN) SIMPLEX P (Stryker Corporation, Kalamazoo, Ml), PALACOS (Smith & Nephew Corporation, United Kingdom), and ENDURANCE (Johnson & Johnson, Inc., New Brunswick, NJ); (g) surgical adhesives containing cyanoacrylates such as DERMABOND (Johnson & Johnson, Inc., New Brunswick, NJ), INDERMIL (U.S. Surgical Company, Norwalk, CT), GLUSTITCH (Blacklock Medical Products Inc., Canada), TlSSUMEND (Veterinary Products Laboratories, Phoenix, AZ), VETBOND (3M Company, St. Paul, MN), HISTOACRYL BLUE (Davis & Geek, St. Louis, MO) and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT (Colgate-Palmolive Company, New York, NY), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the detector/sensor surface); (h) implants containing hydroxyapatite (or synthetic bone material such as calcium sulfate, VITOSS and CORTOSS (both available from Orthovita, Inc., Malvern, PA)) loaded with a fibrosis-inhibiting agent applied to the implantation site (or the detector/sensor surface); (i) other biocompatible tissue fillers alone, or loaded with a fibrosis-inhibiting agent, such as those made by BioCure, Inc. (Norcross, GA), 3M Company and Neomend, Inc. (Sunnyvale, CA), applied to the implantation site (or the detector/sensor surface); (j) polysaccharide gels such as the ADCON series of gels (available from Gliatech, Inc., Cleveland, OH) either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the detector/sensor surface); and/or (k) films, sponges or meshes such as INTERCEED (Gynecare Worldwide, a division of Ethicon, Inc., Somerville, NJ), VICRYL mesh (Ethicon, Inc.), and GELFOAM (Pfizer, Inc., New York, NY) alone, or loaded with a fibrosis-inhibiting agent applied to the implantation site (or the detector/sensor surface).
A preferred polymeric matrix which can be used to help prevent the formation of fibrous tissue on the sensor and/or fibrous encapsulation of the implanted sensor, either alone or in combination with a fibrosis inhibiting agent/composition, is formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydrylj (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue around the implanted sensor.
As should be apparent to one of skill in the art, potentially any anti-scarring agent described below may be utilized alone, or in combination, in the practice of this embodiment. As sensor devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Regardless of the method of application of the drug to the device {i.e., as a coating, incorporated into the structural components of the sensor, or infiltrated into the surrounding tissue), the fibrosis-inhibiting agents, used alone or in combination, may be administered under the following dosing guidelines:
Drugs and dosage: Therapeutic agents that may be used include but are not limited to: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with compositions for treating or preventing surgical adhesions in accordance with the invention. (A)
Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP- 23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10" 8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin Antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10" 8- 10'4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF Kappa B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10" 8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation Factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10'8- 10~4 M of agent should to be maintained on the implant or barrier surface.
2. Implantable Pumps
In another aspect, implantable pumps that include an anti-scarring agent are provided that can be used to deliver drugs to a desired location. Implantable drug delivery devices and pumps are a means to provide prolonged, site-specific release of a therapeutic agent for the management of a variety of medical conditions. Drug delivery implants and pumps are generally utilized when a localized pharmaceutical impact is desired (i.e., the condition affects only a specific region) or when systemic delivery of the agent is inefficient or ineffective (Ae., leads to toxicity or severe side effects, results in inactivation of the drug prior to reaching the target tissue, produces poor symptom/disease control, and/or leads to addiction to the medication). Implantable pumps can also deliver systemic drug levels in a constant, regulated manner for extended periods and help patients avoid the "peaks and valleys" of blood-level drug concentrations associated with intermittent systemic dosing. Another advantage of implantable pumps is improved patient compliance. Many patients forget to take their medications regularly (particularly the young, elderly, chronically ill, mentally handicapped), but with an implantable pump, this problem is alleviated. For many patients this can lead to better symptom control (the dosage can often be titrated to the severity of the symptoms), superior disease management (particularly for insulin delivery in diabetics), and lower drug requirements (particularly for pain medications). Innumerable drug delivery implants and pumps have been used in a variety of clinical applications, including programmable insulin pumps for the treatment of diabetes, intrathecal (in the spine) pumps to administer narcotics (e.g., morphine, fentanyl) for the relief of pain (e.g., cancer, back problems, HIV, post-surgery), local and systemic delivery of chemotherapy for the treatment of cancer (e.g., hepatic artery 5-FU infusion for liver tumors), medications for the treatment of cardiac conditions (e.g., anti-arrhythmic drugs for cardiac rhythm abnormalities), intrathecal delivery of anti-spasmotic drugs (e.g., baclofen) for spasticity in neurological disorders (e.g., Multiple Sclerosis, spinal cord injuries, brain injury, cerebral palsy), or local/regional antibiotics for infection management (e.g., osteomyelitis, septic arthritis). Typically, drug delivery pumps are implanted subcutaneously and consist of a pump unit with a drug reservoir and a flexible catheter through which the drug is delivered to the target tissue. The pump stores and releases prescribed amounts of medication via the catheter to achieve therapeutic drug levels either locally or systemically (depending upon the application). The center of the pump has a self-sealing access port covered by a septum such that a needle can be inserted percutaneously (through both the skin and the septum) to refill the pump with medication as required. There are generally two types of implantable drug delivery pumps. Constant-rate pumps are usually powered by gas and are designed to dispense drugs under pressure as a continual dosage at a preprogrammed, constant rate. The amount and rate of drug flow and regulated by the length of the catheter used, temperature, and altitude and they are best when unchanging, long-term drug delivery is required. Programmable- rate pumps utilize a battery-powered pump and a constant pressure reservoir to deliver drugs on a periodic basis in a manner that can be programmed by the physician or the patient. For the programmable infusion device, the drug may be delivered in small, discrete doses based on a programmed regimen which can be altered according to an individual's clinical response.
In general, drug delivery pumps are implanted to deliver drug at a regulated dose and may, in certain applications, be used in conjunction with implantable sensors that collect information which is used to regulate drug delivery (often called a "closed loop" system). Implantable drug delivery pumps may function and deliver drug in a variety of ways, which include, but are not limited to: (a) delivering drugs only when changes in the body are detected (e.g., sensor stimulated); (b) delivering drugs as a continuous slow release (e.g., constant flow); (c) delivering drugs at prescribed dosages in a pulsatile manner (e.g., non-constant flow); (d) delivering drugs by programmable means; and (e) delivering drugs through a device that is designed for a specific anatomical site (e.g., intraocular, intrathecal, intraperitoneal, intra-arterial or intracardiac). In addition to delivering drugs in a specific way or to a specific location, drug delivery pumps may also be categorized based on their mechanical delivery technology (e.g., the driving force by which drug delivery occurs). For example, the mechanics for delivering drugs may include, without limitation, osmotic pumps, metering systems, peristaltic (roller) pumps, electronically driven pumps, ocular drug delivery pumps and implants, elastomeric pumps, spring-contraction pumps, gas-driven pumps (e.g., induced by electrolytic cell or chemical reaction), hydraulic pumps, piston-dependent pumps and non-piston-dependent pumps, dispensing chambers, infusion pumps, passive pumps, infusate pumps and osmotically-driven fluid dispensers. The clinical function of an implantable drug delivery device or pump depends upon the device, particularly the catheter or drug-dispensing component(s), being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid) and not becoming encapsulated or obstructed by scar tissue. Unfortunately, in many instances when these devices are implanted in the body, they are subject to a "foreign body" response from the surrounding host tissues as described previously. For implantable pumps, the drug-delivery catheter lumen, catheter tip, dispensing components, or delivery membrane may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Alternatively, the entire pump, the catheter and/or the dispensing components can become encapsulated by scar (i.e., the body "walls off' the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule). Either of these developments may lead to inefficient or incomplete drug flow to the desired target tissues or organs (and loss of clinical benefit), while encapsulation can also lead to local drug accumulation (in the capsule) and additional clinical complications (e.g., local drug toxicity; drug sequestration followed by sudden "dumping" of large amounts of drug into the surrounding tissues). Additionally, the tissue surrounding the implantable pump can be inadvertently damaged from the inflammatory foreign body response leading to loss of function and/or tissue damage (e.g., scar tissue in the spinal canal causing pain or obstructing the flow of cerebrospinal fluid).
Implantable drug delivery pumps that release one or more therapeutic agents for reducing scarring at the device-tissue interface (particularly in and around the drug delivery catheter or drug dispensing components) may help prolong the clinical performance of these devices. Inhibition of fibrosis can make sure that the correct amount of drug is dispensed from the device at the appropriate rate and that potentially toxic drugs do not become sequestered in a fibrous capsule. For devices that include electrical or battery components, not only can fibrosis cause the device to function suboptimally or not at all, it can cause excessive drain on battery life as increased energy is required to overcome the increased resistance imposed by the intervening scar tissue.
Virtually any implantable pump may benefit from the present invention. In one aspect, the drug delivery pump may deliver drugs in a continuous, constant-flow, slow release manner. For example, the drug delivery pump may be a passive pump adapted to provide a constant flow of medication which may be regulated by a pressure sensing chamber and a valve chamber in which the constant flow rate may be changed to a new constant flow rate. See, e.g., U.S. Patent No. 6,589,205. In another aspect, the drug delivery pump may deliver drugs at prescribed dosages in a non-constant flow or pulsatile manner. For example, the drug delivery pump may adapt a regular pump to generate a pulsatile fluid drug flow by continuously filling a chamber and then releasing a valve to provide a bolus pulse of the drug. See, e.g., U.S. Patent No. 6,312,409. In another aspect, the drug delivery pump may be programmed to dispense drug in a very specific manner. For example, the drug delivery pump may be a programmable infusate pump composed of a variable volume infusate chamber, and variable volume control fluid pressure and displacement reservoirs, whereby a fluid flow is sampled by a microprocessor based on the programmed value and adjustments are made accordingly to maintain the programmed fluid flow. See, e.g., U.S. Patent No. 4,443,218.
In another aspect, the drug delivery pump suitable for use in the present invention may be manufactured based on different mechanical technologies (e.g., driving forces) of delivering drugs. For example, the drug delivery pump may be an implant composed of a piston that divides two chambers in which one chamber contains a water-swellable agent and the other chamber contains a leuprolide formulation for delivery. See, e.g., U.S. Patent No. 5,728,396. The drug delivery pump may be a non-cylindrical osmotic pump system that may not rely upon a piston to infuse drug and conforms to the anatomical implant site. See, e.g., U.S. Patent No. 6,464,688. The drug delivery pump may be an osmotically driven fluid dispenser composed of a flexible inner bag that contains the drug composition and a port in which the composition can be delivered. See, e.g., U.S. Patent No. 3,987,790. The drug delivery pump may be a fluid-imbibing delivery implant composed of a compartment with a composition permeable to the passage of fluid and has an extended rigid sleeve to resist transient mechanical forces. See, e.g., U.S.
Patent Nos. 5,234,692 and 5,234,693. The drug delivery pump may be a pump with an isolated hydraulic reservoir, metering device, displacement reservoir, drug reservoir, and drug infusion port that is all contained in a housing apparatus. See, e.g., U.S. Patent No. 6,629,954. The drug delivery pump may be composed of a dispensing chamber that has a dispensing passage and valves that are under compressive force to enable drug to flow in a one-way direction. See, e.g., U.S. Patent No. 6,283,949. The drug delivery pump may be spring-driven based on a spring regulating pressure difference with a variable volume drug chamber. See, e.g., U.S. Patent No. 4,772,263. Other examples of drug delivery pumps are described in, e.g., U.S. Patent Nos. 6,645,176; 6,471,688; 6,283,949; 5,137,727 and 5,112,614. In addition, there are osmotically driven drug delivery pumps that are commercially available and suitable for the practice of the invention. These osmotic pumps include the DUROS Implant and ALZET Osmotic Pump from Alza Corporation (Mountain View, CA), which are used to delivery a wide variety of drugs and other therapeutics through the method of osmosis (see, e.g., U.S. Patent Nos. 6,283,953; 6,270,787; 5,660,847; 5,112,614; 5,030,216 and 4,976,966).
As described above, the drug delivery pump can be combined with an agent that inhibits fibrosis to improve performance of the device. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the device (e.g., the polymers that make up the delivery catheters, the semipermeable membranes etc.). Alternatively, or in addition, the fibrosis-inhibiting agent can be infiltrated into the region around the device-tissue interface. It may be obvious to one of skill in the art that commercial drug delivery pumps not specifically cited as well as next- generation and/or subsequently-developed commercial drug delivery products are to be anticipated and are suitable for use under the present invention.
Several specific drug delivery pumps and treatments will be described in greater detail including:
a. Implantable Insulin Pumps for Diabetes
In one aspect, the drug delivery pump may be an insulin pump. Insulin pumps are used for patients with diabetes to replace the need to control blood glucose levels by daily manual injections of insulin. Precise titration of the dosage and timing of insulin administration is a critical component in the effective management of diabetes. If the insulin dosage is too high, blood glucose levels drop precipitously, resulting in confusion and potentially even loss of consciousness. If insulin dosage is too low, blood glucose levels rise too high, leading to excessive thirst, urination, and changes in metabolism known as ketoacidosis. If the timing of insulin administration is incorrect, blood glucose levels can fluctuate wildly between the two extremes - a situation that is thought to contribute to some of the long-term complications of diabetes such as heart disease, kidney failure, nerve damage and blindness. Since in the extreme, all these conditions can be life threatening, the precise dosing and timing of insulin administration is essential to preventing the short and long-term complications of diabetes.
Implantable pumps automate the administration of insulin and eliminate human errors of dosage and timing that can have long-term health consequences. The pump has the capability to inject insulin regularly, multiple times a day and in small doses into the blood stream, peritoneal cavity or subcutaneous tissue. The pump is refilled with insulin once or twice a month by injection directly into the pump chamber. This reduces the number of externally administered injections the patient must undergo and also allows preprogrammed variable amounts of insulin to be released at different times into the blood stream; a situation which more closely resembles normal pancreas function and minimizes fluctuations in blood glucose levels. The insulin pump may be activated by an externally generated signal after the patient has withdrawn a drop of blood, subjected it to an analysis, and made a determination of the amount of insulin that needs to be delivered. However, the most widely pursued application of this technology is the production of a closed- loop "artificial pancreas" which can continuously detect blood glucose levels (through an implanted sensor) and provide feedback to an implantable pump to modulate the administration of insulin to a diabetic patient.
Numerous types of insulin pumps are suitable for use in the practice of the invention. For example, the drug delivery pump may include both an implantable sensor and a drug delivery pump by being composed of a mass of living cells and an electrical signal that regulates the delivery of glucose or glucagon or insulin. See, e.g., U.S. Patent No. 5,474,552. The drug delivery pump may be composed of a single channel catheter with a sensor which is implanted in a vessel that transmits blood chemistry to a subcutaneously implanted infusion device which then dispenses medication through the catheter. See, e.g., U.S. Patent No. 5,109,850. Commercially available insulin pump devices suitable for the practice of the invention include the MINIMED 2007 Implantable Insulin Pump System from Medtronic MiniMed, Inc. (Northridge, CA). The MINIMED pump delivers insulin into the peritoneal cavity in short, frequent bursts to provide insulin to the body similar to that of the normal pancreas (see, e.g., U.S. Patent Nos. 6,558,345 and 6,461 ,331). The MINIMED 2001 Implantable Insulin Pump System (Medtronic MiniMed Inc., Northridge, CA) delivers intraperitoneal insulin injections in a pulsatile manner from a negative pressure reservoir. Both these devices feature a long catheter that transports insulin from the subcutaneously implanted pump into the peritoneal cavity. As described above, the peritoneal drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. In the present invention, the insulin delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters. Alternatively, or in addition, the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface.
It may be obvious to one of skill in the art that commercial drug delivery pumps not specifically cited as well as next-generation and/or subsequently-developed commercial drug delivery products are to be anticipated and are suitable for use under the present invention.
b. Intrathecal Drug Delivery Pumps
In another aspect, intrathecal drug delivery pumps combined with a fibrosis-inhibitor can be used to may used to deliver drugs into the spinal cord for pain management and movement disorders.
Chronic pain is one of the most important clinical problems in all of medicine. For example, it is estimated that over 5 million people in the United States are disabled by back pain. The economic cost of chronic back pain is enormous, resulting in over 100 million lost work days annually at an estimated cost of $50-100 billion. The cost of managing pain for oncology patients is thought to approach $12 billion. Chronic pain disables more people than cancer or heart disease and costs the American public more than both cancer and heart disease combined. In addition to the physical consequences, chronic pain has numerous other costs including loss of employment, marital discord, depression and prescription drug addiction. It goes without saying, therefore, that reducing the morbidity and costs associated with persistent pain remains a significant challenge for the healthcare system.
Intractable severe pain resulting from injury, illness, scoliosis, spinal disc degeneration, spinal cord injury, malignancy, arachnoiditis, chronic disease, pain syndromes (e.g., failed back syndrome, complex regional pain syndrome) and other causes is a debilitating and common medical problem. In many patients, the continued use of analgesics, particularly drugs like narcotics, are not a viable solution due to tolerance, loss of effectiveness, and addiction potential. In an effort to combat this, intrathecal drug delivery devices have been developed to treat severe intractable back pain that is resistant to other traditional treatment modalities such as drug therapy, invasive therapy (surgery), or behavioral/lifestyle changes.
Intrathecal drug delivery pumps are designed and used to reduce pain by delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord. Typically, since this therapy delivers pain medication topically to pain receptors contained in the spinal cord that transmit pain sensation directly to the brain, smaller doses of medication are needed to gain relief. Morphine and other narcotics (usually fentanyl and sufentanil) are the most commonly delivered agents and many patients receive superior relief with lower doses than can be achieved with systemic delivery. Intrathecal drug delivery also allows the administration of pain medications (such as Ziconotide; an N-type calcium channel blocker made by Elan Pharmaceuticals) that cannot cross the blood-brain barrier and are thus only effective when administered by this route. Intrathecal pumps are also used in the management of neurological and movement disorders. Baclofen (marketed as Lioresal by Novartis) is an antispasmotic/muscle relaxant used to treat spasticity and improve mobility in patients with Multiple Sclerosis, cystic fibrosis and spinal injuries. This drug has been proven to be more effective and cause fewer side effects when administered into the CSF by an intrathecal drug delivery pump. Efforts are also underway to treat epilepsy, brain tumors, Alzheimer's disease, Parkinson's disease and Amyetropic Lateral Sclerosis (ALS - Lou Gehrig's disease) via intrathecal administration of agents that may be too toxic to deliver systemically or do not cross the blood-brain barrier. For example, trials of intrathecal^ administered recombinant brain-derived neurotrophic factor (r- BDNF made by Amgen) have been undertaken in ALS patients.
An intrathecal drug delivery system consists of an intrathecal drug infusion pump and an intraspinal catheter, both of which are fully implanted. The pump device is implanted under the skin in the abdominal area, just above or below the beltline and can be refilled by percutaneous injection of the drug into the reservoir. The catheter is tunneled under the skin and runs from the pump to the intrathecal space of the spine. When operational, the pump administers prescribed amounts of medication to the cerebrospinal fluid in either a continuous fashion or in a manner than can be controlled by the physician or the patient in response to symptoms.
Numerous types of implantable intrathecal pumps are suitable for use in combination with a fibrosis-inhibiting agent in the practice of the invention. For example, the implantable pump used to deliver medication may be composed of two osmotic pumps with semipermeable membranes configured to deliver up to two drug delivery regimens at different rates, and having a built-in backup drug delivery system whereby the delivery of drug may continue when the primary delivery system reaches the end of its useful life or fails unexpectedly. See, e.g., U.S. Patent No. 6,471 ,688. The implantable pump may be may be composed of a battery-operated pump unit with a drug reservoir, catheter, and electrodes that are implanted in the epidural space of a patient for relief of pain by delivering a liquid pain-relieving agent through the catheter to the desired location. See, e.g., U.S. Patent No. 5,458,631.
Similar drug-delivery pumps have been described for the infusion of agents into regions of the brain to locally affect the excitability of the neurons in the treatment of a variety of chronic neurogenerative diseases (such as those described above for intrathecal delivery). Implantable pumps may be implanted abdominally which then dispenses drug through a catheter that is tunneled from the abdominal implant site, through the neck to an entry site in the head, and then to the localized treatment site within the brain. Pumps that deliver drug to the brain may discharge the drug at a variety of locations, including, but not limited to, anterior thalamus, ventrolateral thalamus, internal segment of the globus pallidus, substantia nigra pars reticulate, subthalamic nucleus, external segment of globus pallidus, and neostriatum. For example, the drug delivery pump may be composed of an implantable pump portion coupled to a catheter for infusing dosages of drug to a predetermined location of the brain when a sensor detects a symptom, such that a neurological disorder (e.g., seizure) may be treated. See, e.g., U.S. Patent No. 5,978,702. The implantable pump may be implanted adjacent to a predetermined infusion site in a brain such that a predetermined dosage of at least one drug capable of altering the level of excitation of neurons of the brain may be infused such that neurodegeneration is prevented and/or treated. See, e.g., U.S. Patent No. 5,735,814. The implantable pump may include a reservoir for the therapeutic agent which is stored between the galea aponeurotica and cranium of a subject whereby drug is then dispensed via pumping action to the desired location. See, e.g., U.S. Patent No. 6,726,678.
There are numerous commercially available implantable, intrathecal drug-delivery systems which are suitable for the practice of the invention. The SYNCHROMED EL Infusion System which is made by Medtronic, Inc. and is indicated for chronic Intrathecal Baclofen Therapy (ITB Therapy) (see, e.g., U.S. Patent Nos. 6,743,204; 6,669,663; 6,635,048; 6,629,954; 6,626,867; 6,102,678; 5,978,702 and 5,820,589) The SYNCHROMED pump is a programmable, battery-operated device that stores and delivers medication based on the programmed dosing regimen. Medtronic, Inc. (Minneapolis, MN) also sells their ISOMED Constant-Flow Infusion System for use in delivering morphine sulfate directly into the intrathecal space as a treatment for chronic pain. Arrow International produces the Model 3000 infusion pump that provides constant-rate administration of agents such as morphine and baclofen into the intrathecal space. Tricumed Medizintechnik GmbH (Kiel, Germany) produces the Archimedes® constant flow implantable infusion pump for intrathecal administration of pain and antispasmotic drugs. Advanced Neuromodulation Systems (Piano, TX) produces the AccuRx® infusion pump for the treatment of pain and neuromuscular disorders. All these devices feature a long catheter that transports the active agent from a subcutaneously implanted pump into the intrathecal space in the spinal cord. As described above, the intrathecal drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Another potential complication with intrathecal drug delivery is the formation of fibrous tissue in the subdural space that can obstruct CSF flow and lead to serious complications (e.g., hydrocephalus, increased intracranial pressure). In the present invention, the drug delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent and/or prevents fibrosis in the surrounding tissue. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters. Alternatively, or in addition, the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface. The adjuvant use of an anti-infective agent as a catheter coating and /or implant, with or without a fibrosis-inhibiting agent, may also be beneficial in the practice of this invention.
It may be obvious to one of skill in the art that commercial intrathecal drug delivery pumps not specifically cited as well as next-generation and/or subsequently-developed commercial drug delivery products are to be anticipated and are suitable for use under the present invention. c. Implantable Drug Delivery Pumps for Chemotherapy In another aspect, the drug delivery pump may be a pump that dispenses a chemotherapeutic drug for the treatment of cancer. Pumps for dispensing a drug for the treatment of cancer are used to deliver chemotherapeutic agents to a local area of the body. Although virtually any malignancy may potentially be treated in this manner {i.e., by infusing drug directly into a solid tumor or into the blood vessels that supply the tumor), current treatments revolve around the management of hepatic (liver) tumors. For example, FUDR (2'-deoxy 5-fluorouridine) is used in the palliative management of adenocarcinoma (colon, breast, stomach) that has metastasized to the liver. In hepatic artery infusion therapy the drug is delivered via an implantable pump into the artery which provides blood supply to the liver. This allows for higher drug concentrations to reach the liver (the drug is not diluted in the blood as may occur in intravenous administration) and prevents clearance by the liver (the drug is metabolized by the liver and may be rapidly cleared from the bloodstream if administered i.v.); both of which allow higher concentrations of the drug to reach the tumor.
Numerous types of implantable pumps are suitable for delivering chemotherapeutic agents in the practice of the invention. For example, the implantable pump may have a dispensing chamber with a dispensing passage and actuator, reservoir housing with reservoir, and septum for refilling the reservoir. See, e.g., U.S. Patent No. 6,283,949. Medtronic, Inc. sells their ISOMED Constant-Flow Infusion System which may be used to deliver chronic intravascular infusion of floxuridine in a fixed flow rate for the treatment of primary or metastatic cancer. Tricumed Medizintechnik GmbH (Kiel, Germany) sells their ARCHIMEDES DC implantable infusion pump specially adapted to deliver chemotherapy in a constant flow rate within the vicinity of a tumor (see, e.g., U.S. Patent Nos. 5,908,414 and 5,769,823). Arrow International produces the Model 3000 infusion pump that provides constant-rate administration of chemotherapeutic agents into a tumor. All these devices feature a catheter that transports the chemotherapeutic agent from a subcutaneously implanted pump directly into the tumor or the artery that supplies a tumor. As described above, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. If placed intravascularly, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by neointimal tissue which may impair the flow of drug into the blood vessel. In the present invention, the drug delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters. Alternatively, or in addition, the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface. The adjuvant use of an anti-infective agent as a catheter coating and /or implant, with or without a fibrosis-inhibiting agent, may also be beneficial in the practice of this invention. It may be obvious to one of skill in the art that commercial chemotherapy delivery pumps and implants not specifically cited as well as next-generation and/or subsequently-developed commercial chemotherapy delivery products are to be anticipated and are suitable for use in the present invention.
d. Drug Delivery Pumps for the Treatment of Heart Disease
In another aspect, the drug delivery pump may be a pump that dispenses a drug for the treatment of heart disease. Pumps for dispensing a drug for the treatment of heart disease may be used to treat conditions including, but not limited to atrial fibrillation and other cardiac rhythm disorders. Atrial fibrillation is a form of heart disease that afflicts millions of people. It is a condition in which the normal coordinated contraction of the heart is disrupted, primarily by abnormal and uncontrolled action of the atria of the heart. Normally, contractions occur in a controlled sequence with the contractions of the other chambers of the heart. When the right atrium fails to contract, contracts out of sequence, or contracts ineffectively, blood flow from the atria to the ventricles is disrupted. Atrial fibrillation can cause weakness, shortness of breath, angina, lightheadedness and other symptoms due to reduced ventricular filling and reduced cardiac output. Stroke can occur as a result of clot forming in a poorly contracting atria, breaking loose, and traveling via the bloodstream to the arteries of the brain where they become wedged and obstruct blood flow (which may lead to brain damage and death). Typically, atrial fibrillation is treated by medical or electrical conversion (defibrillation), however, complications may exist whereby the therapy causes substantial pain or has the potential to initiate a life threatening ventricular arrhythmia. The pain associated with the electrical shock is severe and unacceptable for many patients, since they are conscious and alert when the device delivers electrical therapy. Medical therapy involves the delivery of anti-arrhythmic drugs by injecting them intravenously, administering them orally or delivering them locally via a drug delivery pump. Numerous types of implantable pumps are described for dispensing a drug for the treatment of heart disease and are suitable for use in the practice of the invention. For example, the drug delivery pump may be an implantable cardiac electrode which delivers stimulation energy and dispenses drug adjacent to the stimulation site. See, e.g., U.S. Patent No. 5,496,360. The drug delivery pump may have a plurality of silicone septii to facilitate the filling of drug reservoirs within the pump which is subcutaneously implanted with a catheter which travels transvenously by way of the subclavian vein through the superior vena cava and into the right atrium for drug delivery. See, e.g., U.S. Patent No. 6,296,630. As described above, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. If placed intravascularly, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by neointimal tissue which may impair the flow of drug into the blood vessel or the right atrium. In the present invention, the drug delivery catheter can be combined with an agent that inhibits fibrosis to keep the delivery catheter lumen patent. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the delivery catheters. Alternatively, or in addition, the fibrosis-inhibiting agent may be infiltrated into the region around the device-tissue interface. The adjuvant use of an anti-infective agent as a catheter coating and /or implant, with or without a fibrosis-inhibiting agent, may also be beneficial in the practice of this invention.
It may be obvious to one of skill in the art that commercial cardiac drug delivery pumps not specifically cited as well as next-generation and/or subsequently-developed commercial cardiac drug delivery products are to be anticipated and are suitable for use under the present invention.
e. Other Drug Delivery Implants Several other implantable pumps have been developed for continuous delivery of pharmaceutical agents.
For example, Debiotech S.A. (Switzerland) has developed the MIP device which is an implantable piezo-actuated silicon micropump for programmable drug delivery applications. This high-performance micropump is based on a MEMS (Micro-Electro-Mechanical) system which allows it to . maintain a low flow rate. The DUROS sufentanil implant from Durect Corporation (Cupertino, CA) is a titanium cylinder that contains a drug reservoir, and a piston driven by an osmotic engine. The VIADUR (leuprolide acetate) implant available from Alza Corporation (Mountain View, CA) uses the same DUROS implant technology to deliver leuprolide over a 12 month period to reduces testosterone levels for the treatment prostate cancer (see, e.g., U.S. Patent Nos. 6,283,953; 6,270,787; 5,660,847; 5,112,614; 5,030,216 and 4,976,966). Fibrous encapsulation of the device can cause failure in a number of ways including: obstructing the semipermeable membrane (which will impair functioning of the osmotic engine by preventing the flow of fluids into the engine), obstructing the exit port (which will impair drug flow out of the device) and/or complete encapsulation (which will create a microenvironment that prevents drug distribution). Many other drug delivery implants, osmotic pumps and the like suffer from similar problems - fibrous encapsulation prevents the appropriate release of drugs into the surrounding tissues. In the present invention, the drug delivery implant can be combined with an agent that inhibits fibrosis to prevent encapsulation, prevent obstruction of the semipermeable membrane and/or to keep the delivery port patent. Fibrosis-inhibiting agents can also be incorporated into, and released from, the materials that are used to construct the drug delivery implant. Alternatively, or in addition, the fibrosis- inhibiting agent may be infiltrated into the tissue around the drug delivery implant.
Although numerous implantable pumps have been described above, all possess similar design features and cause similar unwanted fibrous tissue reactions following implantation. The clinical function of an implantable drug delivery device or pump depends upon the device, particularly the catheter or drug-dispensing component(s), being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid) and not becoming encapsulated or obstructed by scar tissue. For implantable pumps, the drug- delivery catheter lumen, catheter tip, dispensing components, or delivery membrane may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Alternatively, the entire pump, the catheter and/or the dispensing components can become encapsulated by scar (i.e., the body "walls off' the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule). Either of these developments may lead to inefficient or incomplete drug flow to the desired target tissues or organs (and loss of clinical benefit), while encapsulation can also lead to local drug accumulation (in the capsule) and additional clinical complications (e.g., local drug toxicity; drug sequestration followed by sudden "dumping" of large amounts of drug into the surrounding tissues). For implantable pumps that include electrical or battery components, not only can fibrosis cause the device to function suboptimally or not at all, it can cause excessive drain on battery life as increased energy is required to overcome the increased resistance imposed by the intervening scar tissue. Implantable pumps that release a therapeutic agent for reducing scarring at the device-tissue interface can be used to increase efficacy, prolong clinical performance, ensure that the correct amount of drug is dispensed from the device at the appropriate rate, and reduce the risk that potentially toxic drugs become sequestered in a fibrous capsule. In one aspect, the present invention provides implantable pumps that include a fibrosis-inhibiting agent or a composition that includes a fibrosis-inhibiting agent. Numerous polymeric and non-polymeric delivery systems for use in implantable pumps have been described above. These compositions can further include one or more fibrosis- inhibiting agents such that the overgrowth of granulation or fibrous tissue is inhibited or reduced.
Methods for incorporating fibrosis-inhibiting compositions onto or into implantable drug delivery pumps to reduce scarring at the device-tissue interface (particularly in and around the drug delivery catheter or drug dispensing components) include: (a) directly affixing to the implantable pump, catheter and/or drug dispensing components a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier), (b) directly incorporating into the implantable pump, catheter and/or drug dispensing components a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described below, with or without a carrier (c) by coating the implantable pump, catheter and/or drug dispensing components with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the implantable pump, catheter and/or drug dispensing component structure, (e) by inserting the implantable pump, catheter and/or drug dispensing components into a sleeve or mesh which is comprised of, or coated with, a fibrosis-inhibiting composition, (f) constructing the implantable pump itself (or all, or a portion of the catheter and/or drug dispensing components) from a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the implantable pump, catheter and/or drug dispensing component surface, or to a linker (small molecule or polymer) that is coated or attached to the device surface. Each of these methods illustrates an approach for combining an implantable pump with a fibrosis-inhibiting (also referred to herein as anti- scarring) agent according to the present invention.
For implantable pump, the coating process can be performed in such a manner as to: (a) coat a portion of the device (such as the catheter, drug delivery port, semipermeable membrane); or (b) coat the entire device with the fibrosis-inhibiting composition. In addition to, or alternatively, the fibrosis- inhibiting agent can be mixed with the materials that are used to make the implantable pump such that the fibrosis-inhibiting agent is incorporated into the final product. In these manners, a medical device may be prepared which has a coating, where the coating is, e.g., uniform, non-uniform, continuous, discontinuous, or patterned.
In another aspect, an implantable drug delivery pump device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug (i.e., one or more fibrosis-inhibiting agents). The reservoirs may be formed from divets in the device surface or micropores or channels in the device body. In one aspect, the reservoirs are formed from voids in the structure of the device. The reservoirs may house a single type of drug (e.g., fibrosis-inhibiting agent) or more than one type of drug (e.g., a fibrosis-inhibiting agent and an anti-infective agent). The drug(s) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs. The filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier. In certain embodiments, the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount (dose) of drug, and each layer may have a different composition to further tailor the amount and type of drug that is released from the substrate. The multi-layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void. Thus, the coating of the medical device may directly contact the pump, or it may indirectly contact the pump when there is something, e.g., a polymer layer, that is interposed between the pump and the coating that contains the fibrosis-inhibiting agent.
In addition to (or as an alternative to) incorporating a fibrosis- inhibiting agent onto, or into, the implantable pump, catheter and/or drug dispensing components, the fibrosis-inhibiting agent can be applied directly or indirectly to the tissue adjacent to the implantable pump (preferably near in the tissue adjacent to where the drug is delivered from the device). This can be accomplished by applying the fibrosis-inhibiting agent, with or without a polymeric, non-polymeric, or secondary carrier: (a) to the implantable pump, catheter and/or drug dispensing component surface (e.g., as an injectable, paste, gel, or mesh) during the implantation procedure; (b) to the surface of the tissue {e.g., as an injectable, paste, gel, in situ forming gel, or mesh) prior to, immediately prior to, or during, implantation of the implantable pump, catheter and/or drug dispensing components; (c) to the surface of the implantable pump, catheter and/or drug dispensing components and/or to the tissue surrounding the implanted pump, catheter and/or drug dispensing components (e.g., as an injectable, paste, gel, in situ forming gel, or mesh) immediately after implantation; (d) by topical application of the anti-fibrosis agent into the anatomical space where the implantable pump, catheter and/or drug dispensing components will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the region where the implantable pump, catheter and/or drug dispensing components will be inserted); (e) via percutaneous injection into the tissue surrounding the implantable pump, catheter and/or drug dispensing components as a solution, as an infusate, or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, antiplatelet, and/or anti-infective agents) can also be used.
It may be noted that certain polymeric carriers themselves can help prevent the formation of fibrous tissue around the implanted pump, catheter and/or drug dispensing components. These carriers (described below) are particularly useful for the practice of this embodiment, either alone, or in combination with a fibrosis-inhibiting composition. The following polymeric carriers can be infiltrated (as described in the previous paragraph) into the vicinity of the interface between the implanted pump, catheter and/or drug dispensing components of the device and the tissue and include: (a) sprayable collagen-containing formulations such as COSTASIS and CT3, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (b) sprayable PEG-containing formulations such as COSEAL, FOCALSEAL , SPRAYGEL or DURASEAL, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (d) hyaluronic acid-containing formulations such as RESTYLANE, HYLAFORM, PERLANE, SYNVISC, SEPRAFILM, SEPRACOAT1 loaded with a fibrosis-inhibiting agent applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (e) polymeric gels for surgical implantation such as REPEL or FLOWGEL loaded with a fibrosis-inhibiting agent applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (f) orthopedic "cements" used to hold prostheses and tissues in place loaded with a fibrosis-inhibiting agent applied to the implantation site (or the pump, catheter and/or drug dispensing component surface), such as OSTEOBOND, low viscosity cement (LVC), SIMPLEX P, PALACOS, and ENDURANCE; (g) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE SOOTHE-N- SEAL LIQUID PROTECTANT, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (h) implants containing hydroxyapatite (or synthetic bone material such as calcium sulfate, VITOSS and CORTOSS) loaded with a fibrosis-inhibiting agent applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (i) other biocompatible tissue fillers loaded with a fibrosis-inhibiting agent, such as those made by BioCure, Inc., 3M Company and Neomend, Inc., applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); (j) polysaccharide gels such as the ADCON series of gels either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the pump, catheter and/or drug dispensing component surface); and/or (k) films, sponges or meshes such as INTERCEED, VICRYL mesh, and GELFOAM loaded with a fibrosis-inhibiting agent applied to the implantation site (or the pump, catheter and/or drug dispensing component surface).
A preferred polymeric matrix which can be used to help prevent the formation of fibrous tissue around the implanted pump, catheter and/or drug dispensing components, either alone or in combination with a fibrosis inhibiting agent/composition, is formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue around the implanted pump, catheter and/or drug dispensing components.
It may be apparent to one of skill in the art that potentially any anti-scarring agent described below may be utilized alone, or in combination, in the practice of this embodiment. As implantable pumps and their drug delivery mechanisms (e.g., catheters, ports etc.) are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered can be measured, and appropriate surface concentrations of active drug can be determined. Regardless of the method of application of the drug to the device (i.e., as a coating or infiltrated into the surrounding tissue), the fibrosis-inhibiting agents, used alone or in combination, may be administered under the following dosing guidelines:
Drugs and dosage: Therapeutic agents that may be used include but are not limited to ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with compositions for treating or preventing surgical adhesions in accordance with the invention. (A) Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10'4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP- 23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^ - 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10" 8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin Antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, Humicade, Adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10" 8- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^ - 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (J) NF Kappa B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10" 8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation Factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10'8- 10~4 M of agent should to be maintained on the implant or barrier surface. B. Therapeutic Agents for Use with Implantable Sensor and Drug Delivery Pump Devices
As described previously, numerous therapeutic agents are potentially suitable to inhibit fibrous tissue accumulation around the implantable sensor devices and drug-delivery pumps in the manner described herein. The invention provides for medical devices that include an agent that inhibits this tissue accumulation in the vicinity of the device, i.e., between the medical device and the host into which the medical device is implanted. The agent is therefore effective for this goal, is present in an amount that is effective to achieve this goal, and is present at one or more locations that allow for this goal to be achieved, and the device is designed to allow the beneficial effects of the agent to occur. Also, these therapeutic agents can be used alone, or in combination, to prevent scar tissue build-up in the vicinity of the device-tissue interface in order to improve the clinical performance and longevity of these implants.
Suitable therapeutic agents for use in the invention may be readily identified based upon in vitro and in vivo (animal) models, such as those provided in Examples 34-44, 46, and 54. Agents which inhibit fibrosis can also be identified through in vivo models including inhibition of intimal hyperplasia development in the rat balloon carotid artery model (Examples 37 and 46). The assays set forth in Examples 36 and 44 may be used to determine whether an agent is able to inhibit cell proliferation in fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC50 for inhibition of cell proliferation within a range of about 10"6 to about 10"10 M. In certain embodiments, the agent may have an IC50 for inhibition of cell proliferation of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 40 may be used to determine whether an agent may inhibit migration of fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC50 for inhibition of cell migration within a range of about 10"6 to about 10'9M. In certain embodiments, the agent may have an IC50 for inhibition of fibroblast or smooth muscle cell migration of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. Assays set forth herein may be used to determine whether an agent is able to inhibit inflammatory processes, including nitric oxide production in macrophages (Example 34), and/or TN F-alpha production by macrophages (Example 35), and/or IL-1 beta production by macrophages (Example 41), and/or IL-8 production by macrophages (Example 42), and/or inhibition of MCP- 1 by macrophages (Example 43). In one aspect of the invention, the agent has an IC50 for inhibition of any one of these inflammatory processes within a range of about 10"6 to about 10'10M. In certain embodiments, the agent may have an IC50 for any one of these inflammatory processes of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 38 may be used to determine whether an agent is able to inhibit MMP production. In one aspect of the invention, the agent has an IC50 for inhibition of MMP production within a range of about 10"4 to about 10"8M. . In certain embodiments, the agent may have an IC50 for inhibition of MMP production of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 41 (also known as the CAM assay) may be used to determine whether an agent is able to inhibit angiogenesis. In one aspect of the invention, the agent has an IC50 for inhibition of angiogenesis within a range of about 10'6 to about 10'10M. In certain embodiments, the agent may have an IC50 for inhibition of angiogenesis of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 38 may be used to determine whether an agent is able to inhibit MMP- 1. In one aspect of the invention, the agent has an IC50 for inhibition of MMP-1 within a range of about 10 to about 10"10M. In certain embodiments, the agent may have an IC5O for inhibition of MMP-1 of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. Persons skilled in the art would appreciate that an agent which is useful for the compositions and methods described herein may exhibit different levels of effectiveness in different assays (e.g., effectiveness indicated by IC50 values) and in different animal models described herein and practiced in the art. In certain instances, an agent that is useful may exhibit activity in one assay or animal model or may exhibit activity in more than one, two, or three, etc., different assays and/or animal models. Agents which reduce the formation of surgical adhesions may be identified through in vivo models including, for example, the rabbit surgical adhesions model and the rat caecal sidewall model. These pharmacologically active agents (described below) can then be delivered at appropriate dosages into to the tissue either alone, or via carriers (described herein), to treat the clinical problems described herein.
Numerous therapeutic compounds that may be identified as useful in the present invention including:
1. Adensosine A2A receptor antagonist
In another embodiment, the fibrosis-inhibiting compound is an adensosine A2A receptor antagonist (e.g., Sch-63390 (Schering-Plough) or an A2A receptor antagonists from Almirall-Prodesfarma, SCH-58261 (CAS No. 160098-96-4), or an analogue or derivative thereof).
2. AKT inhibitor
In another embodiment, the fibrosis-inhibiting compound is an AKT inhibitor (e.g., PKB inhibitors from DeveloGen, AKT inhibitors from Array BioPharma, Celgene, Merck & Co, Amphora, NeoGenesis Pharmaceuticals, A- 443654 (Abbott Laboratories), erucylphosphocholine (AEterna Zentaris), KRX- 401 (Keryx), protein kinase B inhibitors from Astex Technology, PX-316 (ProlX), or an analogue or derivative thereof).
3. Alpha 2 lntegrin Antagonist
In another embodiment, the fibrosis-inhibiting compound is an alpha 2 integrin antagonist (e.g., Pharmaprojects No. 5754 (Merck KGaA), or an analogue or derivative thereof). 4. Alpha 4 Integrin Antagonist
In another embodiment, the fibrosis-inhibiting compound is an alpha 4 integrin antagonist (e.g., T-0047 (Tanabe Seiyaku), VLA-4 antagonists from Sanofi-Aventis, Merck & Co., Biogen Idee, Uriach, and Molecumetics, alpha 4 integrin antagonists from Genentech), BIO-2421 (Biogen Idee), cell adhesion inhibitors from Kaken Pharmaceuticals, CT-737 (Wyeth), CT-767 (Elan), CY-9652 (Epimmune), CY-9701 (Epimmune), fibronectin antagonists from Uriach, integrin alpha4β7 antagonists frin Wilex, Pharmaprojects No. 5972 (UCB), Pharmaprojects No. 6603 (Wyeth), TBC-3342, TBC-772, and TBC-3486 (Encysive Pharmaceuticals), TBC-4746 (Schering-Plough), or a VLA4Λ/CAM inhibitor (Elan Pharmaceuticals), ZD-7349 (AstraZeneca), or an analogue or derivative thereof).
5. Alpha 7 Nicotinic Receptor Agonist
In another embodiment, the fibrosis-inhibiting compound is an alpha 7 nicotinic receptor agonist (e.g., AZD-0328 (AstraZeneca), galantamine (CAS No. 357-70-0) (Synaptec), MEM-3454 or nicotinic alpha-7 agonist (Memory Pharmaceuticals and Critical Therapeutics), Pharmaprojects No. 4779 (AstraZeneca), PNU-282987 (Pfizer), SSR-180711 (Sanofi-Aventis), TC-1698 or TC-5280 (Targacept), or an analogue or derivative thereof).
6. Angiogenesis Inhibitors
In one embodiment, the fibrosis-inhibiting compound is an angiogenesis inhibitor (e.g., AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC- 4089 (Celgene), HIF-1alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists
(Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT- 116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620
(Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV- 6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (such as combretastatin A-1 , A-2, A-3, A-4, A-5, A-6, B-1 , B-2, B-3, B-4, D-1, D-2, and combretastatin A-4 phosphate (Oxigene)), rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD- 186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC- 68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), or an analogue or derivative thereof). In other embodiments, the angiogenesis inhibitor may be a recombinant anti-angiogenic compound such as ANGIOCOL (available from Biostratum Inc., Durham, NC).
7. Apoptosis Antagonists In another embodiment, the fibrosis-inhibiting compound is an apoptosis antagonist (e.g., didemnin B, RGB-286199 (GPC Biotech), 5F-DF- 203 (Cancer Research Technology), aplidine, bongkrekic acid, triammonium salt, [6]-gingerol (CAS No. 23513-14-6), or an analogue or derivative thereof). 8. Apoptosis Activators
In another embodiment, the fibrosis-inhibiting compound is an apoptosis activator (e.g., aplidine (CAS No. 137219-37-5) (PharmaMar), canfosfamide hydrochloride (CAS No. 58382-37-74 and 39943-59-6) (Telik), idronoxil (CAS No. 81267-65-4) (Novogen), OSI-461 (OSI Pharmaceuticals), DE-098 (Santen), ARQ-550RP (ArQuIe), ABJ-879 (Novartis), adaphostin (NIH), anticancer agents from Apogenix Biotechnology and. Momenta Pharmaceuticals, anti-PARP-1 or anti-PARP-2 (Octamer), BA-1037 (BioAxone), CP-248 (CAS No. 200803-37-8) (OSI Pharmaceuticals), EM-1421 (Erimos), IPI- 504 (Infinity Pharmaceuticals), KP-372-1 (QLT), MPC-6827 (Maxim), MT-103 (Medisyn Technologies), MX-116407 or MX-126374 (Maxim), NPI-0052 (Nereus Pharmaceuticals), NVP-AEW541 (Novartis), PARP inhibitor from Agouron (Pfizer), R-306465 (Johnson & Johnson), TG-100-33 (TargeGen), a XIAP inhibitor from AEgera, ZEN-011 (AEterna Zentaris), canertinib dihydrochloride (CAS No. 289499-45-2) (Pfizer), BH31-1 , 3-BAABE, or an analogue or derivative thereof).
9. Beta 1 Inteqrin Antagonist
In another embodiment, the fibrosis-inhibiting compound is a beta 1 integrin antagonist (e.g., β-1 integrin antagonists, Berkeley Lab, or an analogue or derivative thereof).
10. Beta Tubulin Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a beta tubulin inhibitor (e.g., ZEN-017 (AEterna Zentaris), laulimalide (Kosan Biosciences), or an analogue or derivative thereof).
11. Blockers of Enzyme Production in Hepatitis C
In another embodiment, the fibrosis-inhibiting compound is an agent that blocks enzyme production in hepatitis C (e.g., merimepodib (Vertex Pharmaceuticals), or an analogue or derivative thereof). 12. Bruton's Tyrosine Kinase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a Bruton's tyrosine kinase inhibitor (e.g., a Btk inhibitor from Cellular Genomics, or an analogue or derivative thereof).
13. Calcineurin Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a calcineurin inhibitor (e.g., tacrolimus (LifeCycle Pharma), or an analogue or derivative thereof).
14. Caspase 3 Inhibitors In another embodiment, the fibrosis-inhibiting compound is a caspase 3 inhibitor (e.g., NM-3 (Mercian), or an analogue or derivative thereof).
15. CC Chemokine Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a CC chemokine receptor antagonist (e.g., a chemokine receptor 3 antagonist, a chemokine receptor 6 antagonist, and a chemokine receptor 7 antagonist). Representative examples of CC chemokine receptor antagonists include chemokine antagonists such as the CCR7 antagonists from Neurocrine Biosciences.
In a related embodiment, the fibrosis-inhibiting compound is a CC chemokine receptor antagonist (CCR) 1 , 3, & 5 (e.g., peptide T (Advanced lmmuni T), a CCR3 antagonist from GlaxoSmithKline, a chemokine antagonist (Pharmaprojects No. 6322) from Neurocrine Biosciences or Merck & Co., an HIV therapy agent from ReceptoPharm (Nutra Pharma), Pharmaprojects No. 6129 (Sangamo BioSciences), or an analogue or derivative thereof). In certain embodiments, the CCCR antagonist is a CCR2b chemokine receptor antagonist such as RS 102895 (CAS No. 300815-41-2). 16. Cell Cycle Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cell cycle inhibitor (e.g., SNS-595 (Sunesis), homoharringtonine, or an analogue or derivative thereof). In certain embodiments, the cell cycle inhibitor is an anti- microtubule agent (e.g., synthadotin, or an analogue or derivative thereof).
In certain embodiments, cell cycle inhibitor is a microtubule stimulant (e.g., KRX-0403, or an analogue or derivative thereof).
17. Cathepsin B Inhibitor In another embodiment, the fibrosis-inhibiting compound is a cathepsin B inhibitor (e.g., AM-4299A (Asahi Kasei Pharma), BDI-7800 (Biopharmacopae), a cathepsin B inhibitor from Axys (Celera Genomics), MDL- 104903 (CAS No. 180799-56-8) (Sanofi-Aventis), NC-700 (Nippon Chemiphar), Pharmaprojects No. 2332 (Hoffmann-La Roche), Pharmaprojects No. 4884 (Takeda), Pharmaprojects No. 5134 (Nippon Chemiphar), or an analogue or derivative thereof).
18. Cathepsin K Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a cathepsin K inhibitor (e.g., 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof).
19. Cathepsin L Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a cathepsin L Inhibitor (e.g., a cathepsin L inhibitor from Takeda, INPL-022-E10 (Amura Therapeutics), Pharmaprojects No. 5447 (Taiho), or an analogue or derivative thereof). 20. CD40 Antagonists
In another embodiment, the fibrosis-inhibiting compound is a CD40 antagonists {e.g., 5D12 (Chiron), ABI-793 (Novartis), an anticancer antibody from Chiron, anti-CD40 MAb-2 (Kirin Brewery), anti-CD40 (EIi Lilly), anti-CD40L antibody (UCB), a CD40 inhibitor from Apoxis, CD40 ligand inhibitor from Millennium Pharmaceuticals, a CD40/CAP inhibitor from Snow Brand, CGEN-40 (Compugen), CHIR-12.12 (Chiron), Pharmaprojects No. 5163 (Nippon Kayaku), ruplizumab (Biogen Idee), SGN-40 (Seattle Genetics), TNX- 100 (Akzo Nobel), toralizumab (CAS No. 252662-47-8) (Biogen Idee), or an analogue or derivative thereof).
21. Chemokine Receptor Agonists
In another embodiment, the fibrosis-inhibiting compound is a chemokine receptor agonist (e.g., a chemokine agonist from NeuroTarget, or an analogue or derivative thereof).
22. Chvmase inhibitors
In another embodiment, the fibrosis-inhibiting compound is a chymase inhibitor (e.g., BL-3875 (Dainippon), LEX-043 (SuperGen), NK-3201 (CAS No. 204460-24-2) (Nippon Kayaku), or an analogue or derivative thereof).
23. Collagenase (Interstitial) Antagonists In another embodiment, the fibrosis-inhibiting compound is a collagenase (interstitial) antagonist (e.g., IBFB-212543 (IBFB Pharma), Pharmaprojects No. 3762 (Sanofi-Aventis), S-0885 (CAS No. 117517-22-3) (Sanofi-Aventis), SC-40827 (CAS No. 101470-42-2) (Pfizer), or an analogue or derivative thereof).
24. CXCR (2. 4) Antagonists
In another embodiment, the fibrosis-inhibiting compound is a CXCR (2, 4) antagonist (e.g., SB-656933 (GlaxoSmithKline), AMD3100 octahydrochloride (CAS No. 155148-31-5), or an analogue or derivative thereof).
25. Cvclin Dependent Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cyclin dependent kinase (CDK) inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK-1 inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK-2 inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK- 4 inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK-6 inhibitor. Representative examples of cyclin dependent kinase inhibitors include CAK1 inhibitors from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), or an analogue or derivative thereof.
Additional exemplary cyclin dependent protein kinase inhibitors include an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), CGP74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
26. Cvclooxygenase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cyclooxygenase inhibitor (e.g., NS-398 (CAS No. 123653-11-2), ketoprofen, or an analogue or derivative thereof). In some embodiments, the cyclooxygenase inhibitor is a COX-1 inhibitor such as triflusal, or an analogue or derivative thereof). 27. Dihvdroorotate Dehydrogenase Inhibitor (DHFR) Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a DHFR inhibitor (e.g., PDX (Allos Therapeutics), SC12267, sulfamerazine (CAS No. 127-79-7), or an analogue or derivative thereof).
28. Dual Inteqrin Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a dual integrin inhibitor (e.g., R411 (Roche Pharmaceuticals), or an analogue or derivative thereof).
29. Elastase Inhibitors In another embodiment, the fibrosis-inhibiting compound is an elastase inhibitor (e.g., orazipone, depelestat (CAS No. 506433-25-6) (Dyax), AE-3763 (Dainippon), or an analogue or derivative thereof).
30. Elongation Factor-1 Alpha Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an elongation factor-1 alpha inhibitor (e.g., aplidine, or an analogue or derivative thereof).
31. Endothelial Growth Factor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an endothelial growth factor (EGF) antagonist (e.g., neovastat, NM-3 (Mercian), or an analogue or derivative thereof).
32. Endothelial Growth Factor Receptor Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an endothelial growth factor receptor (EGF-R) kinase inhibitor (e.g., sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No. 125697-92-9), a KDR inhibitor from LG Life Sciences, CT-6685 or CT-6729 (UCB), KRN-633 or KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGF-R inhibitor such as SU 1498, a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, or an analogue or derivative thereof).
In another embodiment, the fibrosis-inhibiting compound is an endothelial growth factor receptor 2 kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
33. Endotoxin Antagonists
In another embodiment, the fibrosis-inhibiting compound is an endotoxin antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof).
34. Epothilone and Tubulin Binders In another embodiment, the fibrosis-inhibiting compound is an epothilone or tubulin binder (e.g., ixabepilone (BMS), or an analogue or derivative thereof).
35. Estrogen Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an estrogen receptor antagonist (e.g., ERB-041 (Wyeth), or an analogue or derivative thereof).
36. FGF Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a FGF inhibitor (e.g., IDN-5390 (Indena), or an analogue or derivative thereof). 37. Farnexyl Transferase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an inhibitor of farnexyl transferase (FTI). In certain embodiments, the FTI inhibits the RAS oncogene family. Examples of FTI's include SARASAR (from Schering Corporation, Kenilworth, NJ), or an analogue or derivative thereof.
38. Farnesyltransferase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a farnesyltransferase inhibitor (e.g., A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIlI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), or an analogue or derivative thereof).
39. FLT-3 Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a FLT- 3 kinase inhibitor (e.g. , Amphora, or an analogue or derivative thereof).
40. FGF Receptor Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a FGF receptor kinase inhibitor (e.g., MED-A300 (Gerolymatos), SSR-128129 (Sanofi- Aventis), TBC-2250 (Encysive Pharmaceuticals), XL-999 (Exelixis), or a FGF receptor kinase inhibitor from Paradigm Therapeutics, or an analogue or derivative thereof).
41. Fibrinogen Antagonists
In another embodiment, the fibrosis-inhibiting compound is a fibrinogen antagonist (e.g., AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, or an analogue or derivative thereof). 42. Heat Shock Protein 90 Antagonists
In another embodiment, the fibrosis-inhibiting compound is a heat shock protein 90 antagonist (e.g., SRN-005 (Sirenade), geldanamycin or a derivative thereof, such as NSC-33050 (17-allylaminogeldanamycin; 17-AAG) or ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), radicicol, Humicola fuscoatra (CAS No. 12772-57-5), or an analogue or derivative thereof).
43. Histone Deacetylase Inhibitors In another embodiment, the fibrosis-inhibiting compound is a histone deacetylase inhibitor (e.g., FK228 (Gloucester), trichostatin A from Streptomyces sp. (CAS No. 58880-19-6), or an analogue or derivative thereof).
44. HMGCoA Reductase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an HMGCoA reductase inhibitor (e.g., an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na )CAS No. 143201-11-0), or an analogue or derivative thereof).
45. ICAM Inhibitors In another embodiment, the fibrosis-inhibiting compound is an
ICAM inhibitor (e.g., alicaforsen (CAS No. 185229-68-9) (ISIS Pharmaceuticals), an ICAM-5 modulator (such as ICAM-4 from ICOS), or an analogue or derivative thereof).
46. IL-1 , ICE & IRAK Antagonists In another embodiment, the fibrosis-inhibiting compound is an IL-
1 , ICE & IRAK antagonist (e.g., CJ-14877 or CP-424174 (Pfizer), NF-61 (Negma-Lerads), or an analogue or derivative thereof). 47. IL-2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an IL-2 inhibitor (e.g., AVE 8062 (Sanofi-Aventis), or an analogue or derivative thereof).
48. Immunosuppressants In another embodiment, the fibrosis-inhibiting compound is an immunosuppressant (e.g., teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis),
Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi- Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), or an analogue or derivative thereof).
49. IMPDH (inosine monophosphate)
In another embodiment, the fibrosis-inhibiting compound is IMPDH (inosine monophosphate) (e.g., ribavirin (Hoffmann-La Roche) or an analogue or derivative thereof). 50. Inteqrin Antagonists
In another embodiment, the fibrosis-inhibiting compound is an integrin antagonist (e.g., 683699 from Glaxo Smith Kline, integrin antagonists from Jerina AG (Germany), or an analogue or derivative thereof).
51. lnterleukin Antagonists
In another embodiment, the fibrosis-inhibiting compound is an interleukin antagonist (e.g., dersalazine, or an analogue or derivative thereof).
In another embodiment, the fibrosis-inhibiting compound is an interleukin 1 antagonist (e.g., NPI-1302a-3, or an analogue or derivative thereof).
52. Inhibitors of Type III Receptor Tyrosine Kinases
In another embodiment, the fibrosis-inhibiting compound is an inhibitor of type III receptor tyrosine kinase such as FLT3, PDGRF and c-KIT (e.g., MLN518 (Millenium Pharmaceuticals), or an analogue or derivative thereof).
53. Irreversible Inhibitors of Enzyme Methionine Aminopeptidase Type 2
In another embodiment, the fibrosis-inhibiting compound is an irreversible inhibitor of enzyme methionine aminopeptidase type 2 (e.g., PPI- 2458 (Praecis Pharmaceuticals), or analogue or derivative thereof).
54. Isozvme-Selective Delta Protein Kinase C Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an isozyme-selective delta protein kinase C inhibitor (e.g., KAI-9803 (Kai Pharmaceuticals), or an analogue or derivative thereof). 55. JAK3 Enzyme Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a JAK3 enzyme inhibitor (e.g., CP-690,550 (Pfizer), or an analogue or derivative thereof).
56. JNK Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a JNK inhibitor (e.g., BF-67192 (BioFocus), XG-101 or XG-102 (Xigen), or an analogue or derivative thereof).
57. Kinase Inhibitors In another embodiment, the fibrosis-inhibiting compound is a kinase inhibitor (e.g., a kinase inhibitors from EVOTEC, or an analogue or derivative thereof).
58. Kinesin Antagonist
In another embodiment, the fibrosis-inhibiting compound is a kinesin antagonist (e.g., SB-715992 and an antifungal from Cytokinetics, or an analogue or derivative thereof).
59. Leukotriene Inhibitors and Antagonists
In another embodiment, the fibrosis-inhibiting compound is a leukotriene inhibitor or antagonist (e.g., ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin -beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 or 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel). Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), or RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1) or SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2) or 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), or analogue or derivative thereof).
60. MAP Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MAP kinase inhibitor (e.g., SRN-003-556 (Sirenade), AEG-3482 (AEgera), ARRY- 142886 (Array BioPharma), CDP-146 (UCB) or analogue or derivative thereof).
61. Matrix Metalloproteinase Inhibitors (MMPI)
In another embodiment, the fibrosis-inhibiting compound is a matrix metalloproteinase inhibitor. A variety of MMPI's may be used in the practice of the invention. In one embodiment, the MMPI is a MMP-1 inhibitor. In another embodiment, the MMPI is a MMP-2 inhibitor. In other embodiments, the MMPI is a MMP-4, MMP-5, MMP-6, MMP-7, or MMP-8 inhibitor. Representative examples of MMPI's include glucosamine sulfate, neovastat, GM1489 (CAS No. 170905-75-6), XL784 (EXEL-01370784), TNF-a Protease lnhibitor-1 or 2 (TAPI-1 or TAPI-2), galardin, or an analogue or derivative thereof.
62. MCP- CCR2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MCP-CCR2 inhibitor (e.g., MLN 1202 (Millennium Pharmaceuticals), or an analogue or derivative thereof). 63. mTOR Inhibitor
In another embodiment, the fibrosis-inhibiting compound is an mTOR inhibitor (e.g., temsirolimus (CAS No. 162635-04-3) (Wyeth), or an analogue or derivative thereof).
64. mTOR Kinase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is an mTOR kinase inhibitor (e.g., ABT-578 (Abbott), temsirolimus (Wyeth), AP- 23573 (Ariad), or an analogue or derivative thereof).
65. Microtubule Inhibitors In another embodiment, the fibrosis-inhibiting compound is a microtubule inhibitor (e.g., antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4 or huMy9- 6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098 or IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, dolastatin 15 (CAS No. 123884-00-4) or an analogue or derivative thereof).
In certain embodiments, the microtubule inhibitor is a microtubule polymerization inhibitor such as vincamine, or an analogue or derivative thereof).
66. MIF Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MIF inhibitor (e.g., AVP-13546 (Avanir), an MIF inhibitor from Genzyme, migration stimulation factor D, or an analogue or derivative thereof). 67. MMP (Stromolvsin) Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MMP (stromolysin) inhibitor (e.g., anticancer tetracycline from Tetragenex, rhostatin (BioAxone), TIMP's from Sanofi-Aventis (CAS No. 86102-31-0), and MMP inhibitors form Cognosci and Tetragenex, or an analogue or derivative thereof).
68. Neurokinin (NK) Antagonist
In another embodiment, the fibrosis-inhibiting compound is a neurokinin (NK) antagonist (e.g., anthrotainin (CAS No. 148084-40-6) (Sanofi- Aventis), an IBS thereapeutic such as SLV-332 from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600 or SSR- 241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), or an analogue or derivative thereof).
69. NF kappa B Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a NF kappa B (NFKB) inhibitor (e.g., emodin (CAS No. 518-82-1), AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104- 49-4) (Encore Pharmaceuticals), Bay 11-7085, or an analogue or derivative thereof). 70. Nitric Oxide Agonists
In another embodiment, the fibrosis-inhibiting compound is a nitric oxide agonist (e.g., Acclaim, Angx-1039 or Angx-3227 (Angiogenix), CAS-1609 (CAS No. 158590-73-9) (Sanofi-Aventis), GCI-503 (Spear Therapeutics), HCT- 3012 (CAS No. 163133-43-5) (NicOx), hydralazine + ISDN (NitroMed), isosorbide dinitrate, Diffutab (CAS No. 87-33-2) (Eurand), isosorbide mononitrate (CAS No. 16051-77-7) from AstraZeneca, Schering AGor Schwarz Pharma, LA-419 (Lacer), molsidomine (CAS No. 25717-80-0) (from Takeda and Therabel), NCX-1000, NCX-2057, or NCX-4040 (NicOx), nitric oxide (ProStrakan), nitroglycerin in the form of a nitroglycerin patch, such as DERMATRANS from (Rottapharm), nitroglycerin (CAS No. 55-63-0) (from Cellegy Pharmaceuticals, Forest Laboratories, NovaDel, Schwarz Pharma, and Watson), NO-releasing prodrugs (Inotek), OM-294DP (OM PHARMA), oxdralazine (CAS No. 27464-23-9) (Sanofi-Aventis), pirsidomine (CAS No. 132722-74-8) (Sanofi-Aventis), prostaglandin and NO donor (Cellegy
Pharmaceuticals), upidosin derivatives (Recordati), or an analogue or derivative thereof).
71. Ornithine Decarboxylase lnhibitiors
In another embodiment, the fibrosis-inhibiting compound is an ornithine decarboxylase inhibitor (e.g., aplidine, or an analogue or derivative thereof).
72. p38 MAP Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a p38 MAP kinase inhibitor (e.g., AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74-6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), or an analogue or derivative thereof). 73. Palmitoyl-Protein Thioesterase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a palmitoyl-protein thioesterase inhibitor (e.g., aplidine, or an analogue or derivative thereof)-
74. PDGF Receptor Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a PDGF receptor kinase inhibitors (e.g., AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), or an analogue or derivative thereof).
75. Peroxisome Proliferator-Activated Receptor Agonists In another embodiment, the fibrosis-inhibiting compound is a peroxisome proliferator-activated receptor (PPAR) agonists (e.g., (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, or AZD-8677 (AstraZeneca), DRF-10945 or balaglitazone (Dr Reddy's), CS-00088 or CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 or MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4) (such as ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), such as AVANDARYL or rosiglitazone maleate/metformin extend (CAS No. 155141-29- 0 and 657-24-9) such as AVANDAMET, or rosiglitazone maleate+metformin, such as AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), or an analogue or derivative thereof).
In certain embodiments, the PPAR Agonist is a PPARα agonist such as GW7647 or fenofibric acid (CAS No. 42017-89-0), a PPAR γ agonist such as MCC-555 (CAS No. 161600-01 -7), GW9662 or GW1929, a PPAERδ agonist such as GW501516, a PPARβ and PPARδ agonist such L-165,041 (CAS No. 79558-09-1), or an analogue or derivative thereof.
76. Phosphatase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a phosphatase inhibitor (e.g., diabetes thereapy such as SQMO3, SQDM38, SQDMδO from Sequenom, Pharmaprojects No. 4191 (Sanofi-Aventis), PRL-3 inhibitors from Genzyme, WIP1 inhibitors from Amgen, or an analogue or derivative thereof).
77. Phosphodiesterase (PDE) Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a phosphodiesterase (PDE) inhibitor (e.g., avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5) or DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, or GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, or IBFB- 211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296- 61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), or an analogue or derivative thereof). In one embodiment, the phosphodiesterase inhibitor is a phosphodiesterase III inhibitor (e.g., enoximone, or an analogue or derivative thereof). In other embodiments, the phosphodiesterase inhibitor is a phosphodiesterase IV inhibitor (e.g., fosfosal, Atopik (Barrier Therapeutics), triflusal, or an analogue or derivative thereof). In other embodiments, the phosphodiesterase inhibitor is a phosphodiesterase V inhibitor.
78. PKC Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a PKC inhibitor (e.g., HMR-105509 or P-10050 (Sanofi-Aventis), JNJ-10164830 (Johnson & Johnson),Ro-31-8425 (CAS No. 131848-97-0),NPC-15437 dihydrochloride (CAS No. 136449-85-9), or an analogue or derivative thereof).
In one embodiment, the PKC inhibitor is an inhibitor of PKC beta (e.g., ruboxistaurin (EIi Lilly), or an analogue or derivative thereof).
79. Platelet Activating Factor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a platelet activating factor antagonist (e.g., dersalazine, or an analogue or derivative thereof). 80. Platelet-Derived Growth Factor Receptor Kinase Inhibitors In another embodiment, the fibrosis-inhibiting compound is a platelet-derived growth factor receptor kinase inhibitor (e.g., sorafenib tosylate, Raf or Ras inhibitors such as sorafenib tosylate from Bayer and Onyx Pharmaceuticals, or an analogue or derivative thereof).
81. Prolyl Hydroxylase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a prolyl hydroxylase inhibitor (e.g., FG-2216 (CAS No. 11096-26-7) or HIF agonists from FibroGen, or an analogue or derivative thereof).
82. Polymorphonuclear Neutrophil Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a polymorphonuclear neutrophil inhibitor (e.g., orazipone, or an analogue or derivative thereof).
83. Protein Kinase B Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a protein kinase B inhibitor (e.g., Akt-1 inhibitors from Amphora, or an analogue or derivative thereof).
84. Protein Kinase C Stimulants In another embodiment, the fibrosis-inhibiting compound is a protein kinase C stimulant (e.g., bryostatin-1 , or analogue or derivative thereof).
85. Purine Nucleoside Analogues
In another embodiment, the fibrosis-inhibiting compound is a purine nucleoside analogue (e.g., cladrinbine and formulations thereof, such as MYLINAX from Serone SA and IVAX Research Inc. (Miami, FL), or an analogue or derivative thereof). 86. Purinoreceptor P2X Antagonist
In another embodiment, the fibrosis-inhibiting compound is a purinoreceptor P2X antagonist (e.g., AZD-9056 (AstraZeneca), R-1554 (Hoffmann-La Roche), AR-C118925XX (AstraZeneca), suramin (CAS No. 129- 46-4), P2Y4 receptor from Euroscreen, or an analogue or derivative thereof).
87. Raf Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a Raf kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
88. Reversible Inhibitors of ErbB1 and ERbB2 In another embodiment, the fibrosis-inhibiting compound is a reversible inhibitor (e.g., lapatinib (GSK), or an analogue or derivative thereof).
89. Ribonucleoside Triphosphate Reductase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cytoplasmic tyrosine kinase inhibitor such as a SRC inhibitor (e.g., SRN-004 (Sirenade), gallium maltolate (Titan Pharmaceutcals), or an analogue or derivative thereof), or an analogue or derivative thereof).
90. SDF-1 Antagonists
In another embodiment, the fibrosis-inhibiting compound is a SDF-1 antagonist (e.g., CTCE-9908 (Chemokine Therapeutics), or an analogue or derivative thereof).
91. Sheddase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a sheddase inhibitor (e.g., INCB-7839 (Incyte Corporation), or an analogue or derivative thereof). 92. SRC Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a SRC inhibitor (e.g., SRN-004 (Sirenade), or an analogue or derivative thereof). In certain embodiments, the SRC inhibitor is a SRC kinase inhibitor (e.g., AZD0530 (AstraZeneca), or an analogue or derivative thereof).
93. Stromelvsin Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a stromelysin inhibitor (e.g., glucosamine sulfate, or an analogue or derivative thereof).
94. Svk Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a syk kinase inhibitor (e.g., R406 (Rigel), or an analogue or derivative thereof).
95. Telomerase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a telomerase inhibitor (e.g., AS-1410 (Antisoma), or an analogue or derivative thereof).
96. TGF Beta Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a TGF beta inhibitor (e.g., pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists (e.g., 1090 and 1091 from Sydney; non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, or an analogue or derivative thereof). 97. TNFα Antagonists and TACE Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a TNFα antagonist or TACE inhibitors (e.g., adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57-7/188913- 58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130- 73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (from Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, or an analogue or derivative thereof).
98. Tumor Necrosis Factor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a tumor necrosis factor (TNF) antagonist (e.g., anti-inflammatory compounds from Biota Inc., or an analogue or derivative thereof).
99. Toll Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a Toll receptor antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof).
100. Tubulin Antagonist
In another embodiment, the fibrosis-inhibiting compound is a tubulin antagonist (e.g., synthadotin, KRX-0403 (Keryx Biopharmaceuticals), or an analogue or derivative thereof).
101. Tyrosine Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a tyrosine kinase inhibitor (e.g., SU-011248 (e.g., SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), (e.g., AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics),
EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax- EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204- 42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50- 1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, or U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL- 647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), herbimycin A (CAS No. 7-563-58-5; AG Scientific; Calbiochem), or an analogue or derivative thereof).
In certain embodiments, the tyrosine kinase inhibitor is an EGFR tyrosine kinase inhibitor such as EKB-569 (Wyeth), or an analogue or derivative thereof).
102. VEGF Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a VEGF Inhibitor (e.g., AZD2171 (AstraZeneca), or an analogue or derivative thereof).
103. Vitamin D Receptor Agonists
In another embodiment, the fibrosis-inhibiting compound is a vitamin D receptor agonist (e.g., BXL-628, BXL-922 (BioXell), or an analogue or derivative thereof).
104) Histamine Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an histamine receptor antagonist. Certain embodiments, the histamine receptor antagonists, such as H1 , H2, and H3 histamine receptor antagonists, block the production of pro-inflammatory cytokines such as TNFa and IL-1 (e.g., IL- 1β). In certain embodiments, the histamine receptor antagonist inhibit NFkB activation. Representative examples of H1 histamine receptor antagonists include phenothiazines, such as promethazine, and alkylamines, such as chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine. Other examples of histamine receptor antagonists include broad spectrum histamine receptor antagonists such as methylxanthines (e.g., theophylline, theobromine, and caffeine). Representative examples of H2 receptor antagonists include those with a histamine-like structure including cimetidine (available under the tradename TAGAMET from SmithKline Beecham Pbamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ)1 famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1). Additional examples include H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt) and antihistamines such as tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones.
105) Alpha Adrenergic Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an alpha adrenergic receptor antagonist. Alpha adrenergic receptor antagonists may inhibit the production of pro-inflammatory cytokines such as TNFa. The alpha adrenergic receptor antagonist may be an alpha-1 and/or an alpha-2 adrenergic receptor antagonist. Representative examples of alpha-1/alpha-2 antagonists include phenoxybenzamine. In certain embodiments, the alpha adrenergic receptor antagonist is a haloalkylamine compound or a catecholamine uptake inhibitor. Representative examples of alpha-1 adrenergic receptor antagonists include phenoxybenzamine hydrochloride and prazosin, a piperizinyl quinazoline. Representative examples of alpha-2 adrenergic receptor antagonists include imadazole based compounds such as idazoxan (CAS No. 79944-56-2), idazoxan hydrochloride, and loxapine succinate salt (CAS No. 27833-64-3). Additional examples of alpha adrenergic receptor antagonists include prazosin hydrochloride.
106) Anti-Psvchotic Compounds
In another embodiment, the fibrosis-inhibiting compound is an anti-psychotic compound, such as a phenothiazine compound or an analogue or derivative thereof. In some embodiments, the fibrosis-inhibiting compound is a phenothiazine derivative capable of suppressing the production of proinflammatory cytokines such as TNFa and/or IL-I . Representative examples of phenothiazine compounds include chlorpromazine, fluphenazine, trifluorphenazine, mesoridazine, thioridazine, and perphenazine. Other examples of anti-psychotic compounds include thioxanthines such as chlorprothixene and thiothixene, clozapine, loxapine succinate, and olanzapine.
107) CaM Kinase Il Inhibitor
In another embodiment, the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof.
108) CaM Kinase Il Inhibitor
In another embodiment, the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof.
109) G Protein Agonist In another embodiment, the fibrosis-inhibiting compound is G protein agonist, such as aluminum fluoride, or an analogue or derivative thereof.
110) Antibiotics and Anti-Microbials
In another embodiment, the fibrosis-inhibiting compound is an antibiotic, such as apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, or an analogue or derivative thereof.
In another embodiment, the fibrosis-inhibiting compound is an anti-microbial agent, such as brefeldin A (CAS No. 20350-15-6), terbinafine, benzoyl peroxide, pentamidine, ornidazole, tinidazole, ketocanazole, sulconazole nitrate salt, or an analogue or derivative thereof. 111) DNA Topoisomerase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is DNA topoisomerase I inhibitor, such as β-lapachone (CAS No. 4707-32-8), or an analogue or derivative thereof. In another embodiment, the fibrosis-inhibiting compound is DNA topoisomerase Il inhibitor, such as (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, or an analogue or derivative thereof.
112) Thromboxane A2 Receptor Inhibitor
In another embodiment, the fibrosis-inhibiting compound is thromboxane A2 receptor inhibitor, such as BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), or an analogue or derivative thereof.
113) D2-Dopamine Receptor Antagonist
In another embodiment, the fibrosis-inhibiting compound is a D2 dopamine receptor antagonist, such as clozapine (CAS No. 5786-21-0), mesoridazine benzenesulfonate, or an analogue or derivative thereof.
114) Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, such as juglone (CAS No. 481- 39-0), or an analogue or derivative thereof.
115) Dopamine Antagonists
In another embodiment, the fibrosis-inhibiting compound is a dopamine antagonist, such as thiothixene, thioridazine hydrochloride, or an analogue or derivative thereof. 116) Anesthetics
In another embodiment, the fibrosis-inhibiting compound is an anesthetic compound, such as lidocaine (CAS No. 137-58-6), or an analogue or derivative thereof.
117) Clotting Factors
In another embodiment, the fibrosis-inhibiting compound is a clotting factor, such as menadione (CAS No. 58-27-5), or an analogue or derivative thereof.
118) Lvsyl Hydrolase Inhibitor In another embodiment, the fibrosis-inhibiting compound is a lysyl hydrolase inhibitor, such as minoxidil (CAS No. 38304-91-5), or an analogue or derivative thereof.
119) Muscarinic Receptor Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a muscarinic receptor inhibitor, such as perphenazine (CAS No. 58-39-9), or an analogue or derivative thereof.
120) Superoxide Anion Generator
In another embodiment, the fibrosis-inhibiting compound is a superoxide anion generator, such as plumbagin (CAS No. 481-42-5), or an analogue or derivative thereof.
. 121) Steroids
In another embodiment, the fibrosis-inhibiting compound is a steroid, such as prednisolone 21-acetate (CAS No. 52-21-1), loteprednol etabonate, (CAS No. 82034-46-6), clobetasol propionate, or an analogue or derivative thereof. 122") Anti-Proliferative Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-proliferative agent, such as silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride or tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methy!-L- arginine acetate salt, galardin, halofuginone hydrobromide (HBr), fascaplysin, or an analogue or derivative thereof.
123) Diuretics In another embodiment, the fibrosis-inhibiting compound is a diuretic, such as spironolactone (CAS No. 52-01-7), or an analogue or derivative thereof.
124) Anti-Coagulants
In another embodiment, the fibrosis-inhibiting compound is an anti-coagulant, such as fucoidan from Fucus vesiculosus (CAS No. 9072-19-9), or an analogue or derivative thereof.
125) Cyclic GMP Agonists
In another embodiment, the fibrosis-inhibiting compound is a cyclic GMP agonist, such as sinitrodil (CAS No. 143248-63-9), or an analogue or derivative thereof.
126) Adenylate Cyclase Agonist
In another embodiment, the fibrosis-inhibiting compound is an adenylate cyclase agonist, such as histamine (CAS No. 51-45-6), or an analogue or derivative thereof. 127) Antioxidants
In another embodiment, the fibrosis-inhibiting compound is an antioxidant, such as morpholine, phytic acid dipotassium salt, (-)- epigallocatechin or (-)-epigallocatechin gallate from green tea (CAS Nos. 970- 74-1 and 1257-08-5, respectively), (-)-epigallocatechin gallate (CAS No. 989- 51-5), nobiletin (CAS No. 478-01-3), probucol (CAS No. 23288-49-5), phosphorous acid, hesperetin, L-ascorbyl-2-phosphate, magnesium salt (CAS No. 84309-23-9), catechin, (±)-naringenin (CAS No. 67604-48-2), (-)- epicatechin, 3-hydroxyflavone, (-)-epicatechin gallate, (-)-arctigenin, or an analogue or derivative thereof.
128) Nitric Oxide Synthase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a nitric oxide synthase inhibitor, such as ammonium pyrrolidinedithiocarbamate (CAS No. 5108-96-3), or an analogue or derivative thereof. In another embodiment, the fibrosis-inhibiting compound is a reversible nitric oxide synthase inhibitor, such as NB-methyl-L-arginine acetate salt (L-NMMA) (CAS No. 53308-83-1), or an analogue or derivative thereof.
129) Anti-Neoplastic Agents
In another embodiment, the fibrosis-inhibiting compound is an antineoplastic agent, such as tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine (CAS Number: 4291-63-8; Leustat®, Janssen-Ortho), imatinib mesilate, or an analogue or derivative thereof.
130) DNA Synthesis Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a DNA synthesis inhibitor, such as S-(2-hydroxy-5-nitrobenyl)-6-thioguanosine or uracilfludarabine phosphate (CAS No. 75607-67-9), 6,11-dihydroxy-5,12- naphthacenedione, cladribine (CAS Number: 4291-63-8; Leustat®, Janssen- Ortho), or an analogue or derivative thereof. 131) DNA Alkylating Aαents
In another embodiment, the fibrosis-inhibiting compound is a DNA alkylating agent, such as dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, or an analogue or derivative thereof.
132) DNA Methylation Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a DNA methylation inhibitor, such as decitabine (CAS No. 2353-33-5), or an analogue or derivative thereof.
133) NSAID Agents In another embodiment, the fibrosis-inhibiting compound is a
NSAID agent, such as nabumetone, benzydamine hydrochloride, or an analogue or derivative thereof.
134) Peptidylglvcine Alpha-Hvdroxylating Monooxygenase Inhibitors In another embodiment, the fibrosis-inhibiting compound is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, such as trans- styrylacetic acid, or an analogue or derivative thereof.
135) MEK1/MEK2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MEK1/MEK 2 inhibitor, such as U0126 (CAS No. 109511-58-2), or an analogue or derivative thereof.
136) NO Synthase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an NO synthase inhibitor, such as L-NAME (CAS No. 53308-83-1), NG-Methyl-L- arginine acetate salt, or an analogue or derivative thereof. 137) Retinoic Acid Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is retinoic acid receptor antagonist, such as isotretinoin (CAS No. 4759-48-2), or an analogue or derivative thereof.
138) ACE Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an ACE inhibitor, such as quinapril hydrochloride (CAS No. 85441-61-8), enalapril, or an analogue or derivative thereof.
139) Glvcosylation Inhibitors In another embodiment, the fibrosis-inhibiting compound is a glycosylation inhibitor, such as aminoguanidine hydrochloride, castanospermine, or an analogue or derivative thereof.
140) Intracellular Calcium Influx Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an intracellular calcium influx inhibitor, such as TAS-301 (CAS No. 193620-69-8), or an analogue or derivative thereof.
141) Anti-Emetic Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-emetic agent, such as amifostine (CAS No. 20537-88-6), or an analogue or derivative thereof.
142) Acetylcholinesterase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an acetylcholinesterase inhibitor, such as (-)-huperzine A (CAS No. 102518-79-6), or an analogue or derivative thereof. 143) ALK-5 Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an ALK-5 receptor antagonist, such as SB 431542 (CAS No. 301836-41-9), or an analogue or derivative thereof.
144) RAR/RXR Antagonists
In another embodiment, the fibrosis-inhibiting compound is a RAR/RXT antagonist, such as 9-cis-retinoic acid, or an analogue or derivative thereof.
145) EIF-2a Inhibitors In another embodiment, the fibrosis-inhibiting compound is a elF-
2a inhibitor, such as salubrinal, or an analogue or derivative thereof.
146) S-Adenosyl-L-Homocvsteine Hydrolase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a S- adenosyl-L-homocysteine hydrolase inhibitor, such as 3-deazaadenosine, or an analogue or derivative thereof.
147) Estrogen Agonists
In another embodiment, the fibrosis-inhibiting compound is an estrogen agonist, such as coumestrol, bisphenol A, 1-Iinoleoyl-rac-glycerol (CAS No. 2277-28-3), daidzein (4,7-dihydroxy-iso-flavone), dihexyl phthalate, kaempferol, formononetin, , or an analogue or derivative thereof.
148) Serotonin Receptor Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a serotonin receptor inhibitor, such as amitriptyline hydrochloride, or an analogue or derivative thereof. 149) Anti-Thrombotic Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-thrombotic agent, such as geniposidic acid, geniposide, or an analogue or derivative thereof.
150) Trvptase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a tryptase inhibitors, such as 2-azetidinone, or an analogue or derivative thereof.
151) Pesticides
In another embodiment, the fibrosis-inhibiting compound is a pesticide, such as allyl disulfide, or an analogue or derivative thereof.
152) Bone Mineralization Promotor
In another embodiment, the fibrosis-inhibiting compound is a bone mineralization promotor, such as glycerol 2-phosphate disodium salt hydrate, or an analogue or derivative thereof.
153) Bisphosphonate Compounds
In another embodiment, the fibrosis-inhibiting compound is a bisphosphonate compound, such as risedronate, or an analogue or derivative thereof.
154) Anti-Inflammatory Compounds In another embodiment, the fibrosis-inhibiting compound is an anti-inflammatory compound, such as aucubin, cepharanthine, or an analogue or derivative thereof. 155) DNA Methylation Promotors
In another embodiment, the fibrosis-inhibiting compound is a DNA methylation promotor, such as 5-azacytidine, or an analogue or derivative thereof.
156) Anti-Spasmodic Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-spasmodic agent, such as 2-hydroxy-4,6-dimethoxyacetophenone, or an analogue or derivative thereof.
157) Protein Synthesis Inhibitors In another embodiment, the fibrosis-inhibiting compound is a protein synthesis inhibitor, such as oxytetracycline hydrochloride, or an analogue or derivative thereof.
158) α-Glucosidase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a α- glucosidase inhibitor, such as myricetin (CAS No. 529-44-2), or an analogue or derivative thereof.
159) Calcium Channel Blockers
In another embodiment, the fibrosis-inhibiting compound is a calcium channel blocker, such as verapamil, nitrendipine, or an analogue or derivative thereof.
In another embodiment, the fibrosis-inhibiting compound is a L- type calcium channel blocker, such as nifedipine (CAS No. 21829-25-4), (+)-cis- diltiazem hydrochloride, or an analogue or derivative thereof.
In another embodiment, the fibrosis-inhibiting compound is a T- type calcium channel blocker, such as penfluridol (CAS No. 26864-56-2), or an analogue or derivative thereof. 160) Pyruvate Dehydrogenase Activators
In another embodiment, the fibrosis-inhibiting compound is a pyruvate dehydrogenase activator, such as dichloroacetic acid, or an analogue or derivative thereof.
161) Prostaglandin Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a prostaglandin inhibitor, such as betulinic acid, or an analogue or derivative thereof.
162) Sodium Channel Inhibitors In another embodiment, the fibrosis-inhibiting compound is a sodium channel inhibitor, such as amiloride hydrochloride hydrate, or an analogue or derivative thereof.
163) Serine Protease Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a serine protease inhibitor, such as gabexate mesylate, or an analogue or derivative thereof.
164) Intracellular Calcium Flux Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an intracellular calcium flux inhibitor, such as thapsigargin, or an analogue or derivative thereof.
165) JAK2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a JAK2 inhibitor (e.g., AG-490 (CAS No. 134036-52-5), or an analogue or derivative thereof). 166) Androgen Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an androgen inhibitor (e.g., tibolone (CAS No. 5630-53-5), or an analogue or derivative thereof).
167) Aromatase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an aromatase inhibitor (e.g., letrozole, or an analogue or derivative thereof).
168) Anti-Viral Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-viral agent, such as imiquimod, or an analogue or derivative thereof.
169) 5-HT Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a 5-HT inhibitor, such as ketanserin tartrate, amoxapine, or an analogue or derivative thereof.
170) FXR Antagonists
In another embodiment, the fibrosis-inhibiting compound is a FXR antagonist, such as guggulsterone (CAS No. 95975-55-6), or an analogue or derivative thereof.
171) Actin Polymerization and Stabilization Promotors In another embodiment, the fibrosis-inhibiting compound is an actin polymerization and stabilization promotor, such as jasplakinolide, or an analogue or derivative thereof. 172) AXOR12 Agonists
In another embodiment, the fibrosis-inhibiting compound is an AXOR12 agonist, such as metastin (KiSS-1 (112-121), or an analogue or derivative thereof.
173) Angiotensin Il Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an angiotensin Il receptor agonist, such as losartan potassium, or an analogue or derivative thereof.
174) Platelet Aggregation Inhibitors In another embodiment, the fibrosis-inhibiting compound is a platelet aggregation inhibitor, such as clopidogrel, or an analogue or derivative thereof.
175) CB1/CB2 Receptor Agonists
In another embodiment, the fibrosis-inhibiting compound is a CB1/CB2 receptor agonist, such as HU-210 (CAS No. 112830-95-2), or an analogue or derivative thereof.
176) Norepinephrine Reuptake Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a norepinephrine reuptake inhibitor, such as nortriptyline hydrochloride, or an analogue or derivative thereof.
177) Selective Serotonin Reuptake Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a selective serotonin reuptake inhibitor, such as paroxetine maleate, or an analogue or derivative thereof. 178) Reducing Agents
In another embodiment, the fibrosis-inhibiting compound is a reducing agent such as WW-85 (lnotek), or an analogue or derivative thereof.
179) Immuno-modulators
C. In another embodiment, the fibrosis-inhibiting compound is an immunomodulators such as Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, or an analogue or derivative thereof.Combination Therapies
In addition to incorporation of a fibrosis-inhibiting agent, one or more other pharmaceutically active agents can be incorporated into the present compositions to improve or enhance efficacy. In one aspect, the composition may further include a compound which acts to have an inhibitory effect on pathological processes in or around the treatment site. Representative examples of additional therapeutically active agents include, by way of example and not limitation, anti-thrombotic agents, anti-proliferative agents, antiinflammatory agents, neoplastic agents, enzymes, receptor antagonists or agonists, hormones, antibiotics, antimicrobial agents, antibodies, cytokine inhibitors, IMPDH (inosine monophosplate dehydrogenase) inhibitors tyrosine kinase inhibitors, MMP inhibitors, p38 MAP kinase inhibitors, immunosuppressants, apoptosis antagonists, caspase inhibitors, and JNK inhibitors.
In one aspect, the present invention also provides for the combination of an implantable pump or implantable sensor device (as well as compositions and methods for making implantable pump and sensor devices) that includes an anti-fibrosing agent and an anti-infective agent, which reduces the likelihood of infections.
Infection is a common complication of the implantation of foreign bodies such as, for example, medical devices. Foreign materials provide an ideal site for micro- organisms to attach and colonize. It is also hypothesized that there is an impairment of host defenses to infection in the microenvironment surrounding a foreign material. These factors make medical implants particularly susceptible to infection and make eradication of such an infection difficult, if not impossible, in most cases. The present invention provides agents (e.g., chemotherapeutic agents) that can be released from a composition, and which have potent antimicrobial activity at extremely low doses. A wide variety of anti-infective agents can be utilized in combination with the present compositions. Suitable anti-infective agents may be readily determined based the assays provided in Example 51. Discussed in more detail below are several representative examples of agents that can be used: (A) anthracyclines {e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin).
(A) Anthracvclines
Anthracyclines have the following general structure, where the R groups may be a variety of organic groups:
Figure imgf000137_0001
According to U.S. Patent 5,594,158, suitable R groups are as follows: R1 is CH3 or CH2OH; R2 is daunosamine or H; R3 and R4 are independently one of OH, NO2, NH2, F, Cl, Br, I, CN, H or groups derived from these; R5 is hydrogen, hydroxyl, or methoxy; and R6-S are all hydrogen.
Alternatively, R5 and R6 are hydrogen and R7 and R8 are alkyl or halogen, or vice versa. According to U.S. Patent 5,843,903, R1 may be a conjugated peptide. According to U.S. Patent 4,296,105, R5 may be an ether linked alkyl group. According to U.S. Patent 4,215,062, R5 may be OH or an ether linked alkyl group. Ri may also be linked to the anthracycline ring by a group other than C(O), such as an alkyl or branched alkyl group having the C(O) linking moiety at its end, such as -CH2CH(CH2-X)C(O)-R1, wherein X is H or an alkyl group (see, e.g., U.S. Patent 4,215,062). R2 may alternately be a group linked by the functional group =N-NHC(O)-Y, where Y is a group such as a phenyl or substituted phenyl ring. Alternately R3 may have the following structure:
Figure imgf000138_0001
in which R9 is OH either in or out of the plane of the ring, or is a second sugar moiety such as R3. Ri0 may be H or form a secondary amine with a group such as an aromatic group, saturated or partially saturated 5 or 6 membered heterocyclic having at least one ring nitrogen (see U.S. Patent 5,843,903).
Alternately, Ri0 may be derived from an amino acid, having the structure -
C(O)CH(NHRii)(Ri2), in which Rn is H, or forms a C3-4 membered alkylene with Ri2. Ri2 may be H, alkyl, aminoalkyl, amino, hydroxyl, mercapto, phenyl, benzyl or methylthio (see U.S. Patent 4,296,105).
Exemplary anthracyclines are doxorubicin, daunorubicin, idambicin, epirubicin, pirarubicin, zorubicin, and carubicin. Suitable compounds have the structures:
Figure imgf000138_0002
Ri R2 R3
Doxorubicin: OCH3 C(O)CH2OH OH out of ring plane
Epirubicin: (4' epimer of OCH3 C(O)CH2OH OH in ring plane doxorubicin)
Daunorubicin: OCH3 C(O)CH3 OH out of ring plane
Idarubicin: H C(O)CH3 OH out of ring plane
Pirarubicin: OCH3 C(O)CH2OH < -1
Zorubicin: OCH3 C(CH3X=N)NHC(O)C6H5 OH
Carubicin: OH C(O)CH3 OH out of ring plane
Other suitable anthracyclines are anthramycin, mitoxantrone, menogaril, nogalamycin, aclacinomycin A, olivomycin A, chromomycin A3, and plicamycin having the structures:
R, R2 R5 COOCH3
Figure imgf000139_0001
Figure imgf000139_0002
Olivomycin A COCH(CI-Ij)2 CH3 C(XH3 H
Chramom/cinAj COCH3 CH3 COCH3 CH5
Plicamycin H H H CH3
Figure imgf000139_0003
Other representative anthracyclines include, FCE 23762, a doxorubicin derivative (Quaglia et al., J. Liq. Chromatogr. 17(18):3911-3923, 1994), annamycin (Zou et al., J. Pharm. ScL 82(11):1151-1154, 1993), ruboxyl (Rapoport ef al., J. Controlled Release 58(2): 153- 162, 1999), anthracycline disaccharide doxorubicin analogue (Pratesi et al., Clin. Cancer Res. 4(11):2833-2839, 1998), N-(trifluoroacetyl)doxorubicin and 4'-O-acetyl-N- (trifluoroacetyl)doxorubicin (Berube & Lepage, Synth. Commun. 28(6): 1109- 1116, 1998), 2-pyrrolinodoxorubicin (Nagy et al., Proc. Nat'IAcad. ScL U.S.A. 95(4): 1794-1799, 1998), disaccharide doxorubicin analogues (Arcamone et al., J. Nat'l Cancer Inst. 89(16):1217-1223, 1997), 4-demethoxy-7-O-[2,6-dideoxy- 4-O-(2,3,6-trideoxy-3-amino-α-L-lyxo-hexopyranosyl)-α-L-lyxo-hexopyranosyl]- adriamicinone doxorubicin disaccharide analogue (Monteagudo et a/., Carbohydr. Res. 300(1):11-16, 1997), 2-pyrrolinodoxorubicin (Nagy et al., Proc. Nat'IAcad. ScL U.S.A. 94(2):652-656, 1997), morpholinyl doxorubicin analogues (Duran et al., Cancer Chemother. Pharmacol. 38(3):210-216, 1996), enaminomalonyl-β-alanine doxorubicin derivatives (Seitz et al., Tetrahedron Lett. 36(9): 1413-16, 1995), cephalosporin doxorubicin derivatives (Vrudhula et al., J. Med. Chem. 38(8): 1380-5, 1995), hydroxyrubicin (Solary et al., Int. J. Cancer 58(1 ):85-94, 1994), methoxymorpholino doxorubicin derivative (Kuhl et al., Cancer Chemother. Pharmacol. 33(1):10-16, 1993), (6- maleimidocaproyl)hydrazone doxorubicin derivative (Willner et al., Bioconjugate Chem. 4(6):521-7, 1993), N-(5,5-diacetoxypent-1-yl) doxorubicin (Cherif & Farquhar, J. Med. Chem. 35(17):3208-14, 1992), FCE 23762 methoxymorpholinyl doxorubicin derivative (Ripamonti et al., Br. J. Cancer 65(5):703-7, 1992), N-hydroxysuccinimide ester doxorubicin derivatives (Demant et al., Biochim. Biophys. Acta 1118(1):83-90, 1991), polydeoxynucleotide doxorubicin derivatives (Ruggiero et al., Biochim. Biophys. Acta 1129(3):2Q4-302, 1991), morpholinyl doxorubicin derivatives (EPA 434960), mitoxantrone doxorubicin analogue (Krapcho et al., J. Med. Chem. 34(8):2373-80. 1991), AD198 doxorubicin analogue (Traganos et al., Cancer Res. 57(14):3682-9, 1991), 4-demethoxy-3'-N-trifluoroacetyldoxorubicin (Horton et al., Drug Des. Delivery 6(2): 123-9, 1990), 4'-epidoxorubicin (Drzewoski et a/., Pol. J. Pharmacol. Pharm. 40(2): 159-65, 1988; Weenen et a/., Eur. J. Cancer CHn. Oncol. 20(7):919-26, 1984), alkylating cyanomorpholino doxorubicin derivative (Scudder et al., J. Nat'l Cancer Inst. 80(16): 1294-8, 1988), deoxydihydroiodooxorubicin (EPA 275966), adriblastin (Kalishevskaya et al., Vestn. Mosk. Univ., 16(B\o\. 1):21-7, 1988), 4'-deoxydoxorubicin (Schoelzel et al., Leuk. Res. 70(12): 1455-9, 1986), 4-demethyoxy-4'-o-methyldoxorubicin (Giuliani et ai, Proc. Int. Congr. Chemother. 76:285-70-285-77, 1983), 3'- deamino-3'-hydroxydoxorubicin (Horton et ai, J. Antibiot. 37(8):853-8, 1984), 4- demethyoxy doxorubicin analogues (Barbieri et al., Drugs Exp. CHn. Res. 70(2):85-90, 1984), N-L-leucyl doxorubicin derivatives (Trouet et ai., Anthracyclines (Proc. Int. Symp. Tumor Pharmacother.), 179-81 , 1983), 3'- deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (U.S. 4,314,054), 3'-deamino-3'-(4-mortholinyl) doxorubicin derivatives (U.S. 4,301 ,277), 4'- deoxydoxorubicin and 4'-o-methyldoxorubicin (Giuliani et ai., Int. J. Cancer 27(1):5-13, 1981), aglycone doxorubicin derivatives (Chan & Watson, J. Pharm. Sci. 67(12): 1748-52, 1978), SM 5887 (Pharma Japan 1468:20, 1995), MX-2 (Pharma Japan 1420:19, 1994), 4'-deoxy-13(S)-dihydro-4'-iododoxorubicin (EP 275966), morpholinyl doxorubicin derivatives (EPA 434960), 3'-deamino-3'-(4- methoxy-1-piperidinyl) doxorubicin derivatives (U.S. 4,314,054), doxorubicin- 14-valerate, morpholinodoxorubicin (U.S. 5,004,606), 3'-deamino-3'-(3"-cyano- 4"-morpholinyl doxorubicin; 3'-deamino-3'-(3"-cyano-4"-morpholinyl)-13- dihydoxorubicin; (3'-deamino-3'-(3"-cyano-4"-morpholinyl) daunorubicin; 3'- deamino-3'-(3"-cyano-4"-morpholinyl)-3-dihydrodaunorubicin; and 3'-deamino- 3'-(4"-morpholinyl-5-iminodoxorubicin and derivatives (U.S. 4,585,859), 3'- deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (U.S. 4,314,054) and 3-deamino-3-(4-morpholinyl) doxorubicin derivatives (U.S. 4,301 ,277).
(B) Fluoropyrimidine analogues
In another aspect, the therapeutic agent is a fluoropyrimidine analog, such as 5-fluorouracil, or an analogue or derivative thereof, including carmofur, doxifluridine, emitefur, tegafur, and floxuridine. Exemplary compounds have the structures:
Figure imgf000142_0001
R1 R2
5-Fluorouracil H H
Carmofur C(O)NH(CH2)5CH3 H
Doxifluridine A1 H
Floxuridine A2 H
Emitefur CH2OCH2CH3 B
Tegafur C H
Figure imgf000142_0002
Other suitable fluoropyrimidine analogues include 5-FudR (5- fluoro-deoxyuridine), or an analogue or derivative thereof, including 5- iododeoxyuridine (5-ludR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP). Exemplary compounds have the structures:
Figure imgf000143_0001
5-Fluoro-2'-deoxyuridine: R = F 5-Bromo-2'-deoxyuridine: R = Br 5-lodo-2'-deoxyuridine: R = I
Other representative examples of fluoropyrimidine analogues include N3-alkylated analogues of 5-fluorouracil (Kozai et al., J. Chem. Soc, Perkin Trans. 7(19):3145-3146, 1998), 5-fluorouracil derivatives with 1 ,4- oxaheteroepane moieties (Gomez et al., Tetrahedron 54(43):13295-13312, 1998), 5-fluorouracil and nucleoside analogues (Li, Anticancer Res. 77(1A):21- 27, 1997), cis- and trans-5-fluoro-5,6-dihydro-6-alkoxyuracil (Van der Wilt et al., Br. J. Cancer 68(4)102-1 , 1993), cyclopentane 5-fluorouracil analogues (Hronowski & Szarek, Can. J. Chem. 70(4): 1162-9, 1992), A-OT-fluorouracil (Zhang et al., Zongguo Yiyao Gongye Zazhi 20(11):513-15, 1989), N4- trimethoxybenzoyl-5'-deoxy-5-fluorocytidine and 5'-deoxy-5-fluorouridine (Miwa et al., Chem. Pharm. Bull. 38(4):998-1003, 1990), 1-hexylcarbamoyl-5- fluorouracil (Hoshi et al., J. Pharmacobio-Dun. 3(9):478-81 , 1980; Maehara et al., Chemotherapy (Basel) 34(6):484-9, 1988), B-3839 (Prajda et al., In Vivo 2(2): 151 -4, 1988), uracil-i-^-tetrahydrofuryO-δ-fluorouracil (Anai etal., Oncology 45(3):144-7, 1988), 1-(2'-deoxy-2'-fluoro-β-D-arabinofuranosyl)-5- fluorouracil (Suzuko et al., MoI. Pharmacol. 37(3):301-6, 1987), doxifluridine (Matuura et al. , Oyo Yakuri 29(5):803-31 , 1985), 5'-deoxy-5-fluorouridine
(Bollag & Hartmann, Eur. J. Cancer 16(4) :427-32, 1980), 1-acetyl-3-O-toluyl-5- fluorouracil (Okada, Hiroshima J. Med. Sci. 2δ(1):49-66, 1979), 5-fluorouracil- m-formylbenzene-sulfonate (JP 55059173), N'-(2-furanidyl)-5-fluorouracil (JP 53149985) and 1-(2-tetrahydrofuryl)-5-fluorouracil (JP 52089680). These compounds are believed to function as therapeutic agents by serving as antimetabolites of pyrimidine.
(C) Folic acid antagonists
In another aspect, the therapeutic agent is a folic acid antagonist, such as methotrexate or derivatives or analogues thereof, including edatrexate, trimetrexate, raltitrexed, piritrexim, denopterin, tomudex, and pteropterin. Methotrexate analogues have the following general structure:
Figure imgf000144_0001
The identity of the R group may be selected from organic groups, particularly those groups set forth in U.S. Patent Nos. 5,166,149 and 5,382,582. For example, Ri may be N, R2 may be N or C(CH3), R3 and R3' may H or alkyl, e.g., CH3, R4 may be a single bond or NR, where R is H or alkyl group. R5|6,8 may be H, OCH3, or alternately they can be halogens or hydro groups. R7 is a side chain of the general structure:
Figure imgf000144_0002
wherein n = 1 for methotrexate, n = 3 for pteropterin. The carboxyl groups in the side chain may be esterified or form a salt such as a Zn2+ salt. Rg and R10 can be NH2 or may be alkyl substituted.
Exemplary folic acid antagonist compounds have the structures:
Figure imgf000145_0001
Figure imgf000145_0002
Methotrexate NH2 N N H N(CH3) H H A (n=1) H
Edatrexate NH2 N N H CH(CH2CH3) H H A (n=1) H
Trimetrexate NH2 CH C(CH3) H NH H OCH3 OCH3 OCH3
Pteropterin OH N N H NH H H A (n=3) H
Denopterin OH N N CH3 N(CH3) H H A (n=1) H
Peritrexim NH2 N C(CH3) H single bond OCH3 H H OCH3
Figure imgf000145_0003
Figure imgf000145_0004
Tomudex
Other representative examples include 6-S-aminoacyloxymethyl mercaptopurine derivatives (Harada et al., Chem. Pharm. Bull. 43(10):793-6, 1995), 6-mercaptopurine (6-MP) (Kashida et al., Biol. Pharm. Bull. 78(11):1492- 7, 1995), 7,8-polymethyleneimidazo-1,3,2-diazaphosphorines (Nilov et al., Mendeleev Commun. 2:67, 1995), azathioprine (Chifotides et al., J. Inorg. Biochem. 56(4):249-64, 1994), methyl-D-glucopyranoside mercaptopurine derivatives (Da Silva et al., Eur. J. Med. Chem. 29(2):149-52, 1994) and s- alkynyl mercaptopurine derivatives (Ratsino et al., Khim.-Farm. Zh. 75(8):65-7, 1981); indoline ring and a modified ornithine or glutamic acid-bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 45(7):1146- 1150, 1997), a I ky l-su bstituted benzene ring C bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 44(12):2287-2293, 1996), benzoxazine or benzothiazine moiety-bearing methotrexate derivatives (Matsuoka et al. , J. Med. Chem. 40(1):105-111 , 1997), 10-deazaaminopterin analogues (DeGraw et al., J. Med. Chem. 40(3):370-376, 1997), 5-deazaaminopterin and 5,10- dideazaaminopterin methotrexate analogues (Piper et al., J. Med. Chem. 40(3):377-384, 1997), indoline moiety-bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 44(7): 1332-1337, 1996), lipophilic amide methotrexate derivatives (Pignatello et al., World Meet. Pharm. Biopharm. Pharm. Technol., 563-4, 1995), L-threo-(2S,4S)-4-fluoroglutamic acid and DL- 3,3-difluoroglutamic acid-containing methotrexate analogues (Hart et al., J. Med. Chem. 39(1):56-65, 1996), methotrexate tetrahydroquinazoline analogue (Gangjee, et al., J. Heterocycl. Chem. 32(1):243-8, 1995), N-(α-aminoacyl) methotrexate derivatives (Cheung et al., Pteridines 3( 1-2): 101 -2, 1992), biotin methotrexate derivatives (Fan et al. , Pteridines 3(1 -2): 131 -2, 1992), D-glutamic acid or D-erythrou, threo-4-fluoroglutamic acid methotrexate analogues (McGuire et al., Biochem. Pharmacol. 42(12):2400-3, 1991), β,γ-methano methotrexate analogues (Rosowsky et al., Pteridines 2(3):133-9, 1991), 10- deazaaminopterin (10-EDAM) analogue (Braakhuis et al., Chem. Biol. Pteridines, Proc. Int. Symp. Pteridines Folic Acid Deriv., 1027-30, 1989), γ- tetrazole methotrexate analogue (Kalman et al., Chem. Biol. Pteridines, Proc. Int. Symp. Pteridines Folic Acid Deriv., 1154-7, 1989), N-(L-α-aminoacyl) methotrexate derivatives (Cheung et al., Heterocycles 28(2):751-8, 1989), meta and ortho isomers of aminopterin (Rosowsky et al., J. Med. Chem. 32(12):2582, 1989), hydroxymethylmethotrexate (DE 267495), γ-fluoromethotrexate
(McGuire et al., Cancer Res. 49(16):4517-25, 1989), polyglutamyl methotrexate derivatives (Kumar et al., Cancer Res. 46(10):5020-3, 1986), gem- diphosphonate methotrexate analogues (WO 88/06158), α- and γ-substituted methotrexate analogues (Tsushima et al., Tetrahedron 44(17):5375-87, 1988), 5-methyl-5-deaza methotrexate analogues (4,725,687), Nδ-acyl-Nα-(4-amino-4- deoxypteroyl)-L-omithine derivatives (Rosowsky et al., J. Med. Chem. 37(7):1332-7, 1988), 8-deaza methotrexate analogues (Kuehl et al., Cancer Res. 48(6):1481-8, 1988), acivicin methotrexate analogue (Rosowsky et al., J. Med. Chem. 30(8):1463-9, 1987), polymeric platinol methotrexate derivative (Carraher et al., Polym. Sci. Technol. (Plenum), 35(Adv. Biomed. Po/ym.j:311- 24, 1987), methotrexate-γ-dimyristoylphophatidylethanolamine (Kinsky et al., Biochim. Biophys. Acta 977(2):211-18, 1987), methotrexate polyglutamate analogues (Rosowsky et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 985-8, 1986), poly-γ-glutamyl methotrexate derivatives (Kisliuk et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 989-92, 1986), deoxyuridylate methotrexate derivatives (Webber et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 659-62, 1986), iodoacetyl lysine methotrexate analogue (Delcamp et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 807-9, 1986), 2,.omega.-diaminoalkanoid acid-containing methotrexate analogues (McGuire et al., Biochem. Pharmacol. 35(15):2607-13, 1986), polyglutamate methotrexate derivatives (Kamen & Winick, Methods Enzymol. 722(Vitam. Coenzymes, Pt. G):339-46, 1986), 5-methyl-5-deaza analogues (Piper et al., J. Med. Chem. 29(6): 1080-7, 1986), quinazoline methotrexate analogue (Mastropaolo et al., J. Med. Chem. 29(1):155-8, 1986), pyrazine methotrexate analogue (Lever & Vestal, J. Heterocycl. Chem. 22(1):5-6, 1985), cysteic acid and homocysteic acid methotrexate analogues (4,490,529), γ-tert-butyl methotrexate esters (Rosowsky et al., J. Med. Chem. 28(5):660-7, 1985), fluorinated methotrexate analogues (Tsushima et al., Heterocycles 23(1):45-9, 1985), folate methotrexate analogue (Trombe, J. Bacteriol. 760(3):849-53, 1984), phosphonoglutamic acid analogues (Sturtz & Guillamot, Eur. J. Med. Chem.-Chim. Ther. 79(3):267-73, 1984), poly (L-lysine) methotrexate conjugates (Rosowsky et al., J. Med. Chem. 27(7):888-93, 1984), dilysine and trilysine methotrexate derivates (Forsch & Rosowsky, J. Org. Chem. 49(7):1305-9, 1984), 7-hydroxymethotrexate (Fabre et al., Cancer Res. 43(10):4648-52, 1983), poly-γ-glutamyl methotrexate analogues (Piper & Montgomery, Adv. Exp. Med. Biol., 163(FoIyI Antifolyl Polyglutamates):95--\00, 1983), 3\5'-dichloromethotrexate (Rosowsky & Yu, J. Med. Chem. 26(10):1448- 52, 1983), diazoketone and chloromethylketone methotrexate analogues (Gangjee et al., J. Pharm. Sci. 77(6):717-19, 1982), 10-propargylaminopterin and alkyl methotrexate homologs (Piper et al., J. Med. Chem. 25(7):877-80, 1982), lectin derivatives of methotrexate (Lin et al., JNCI 66(3):523-8, 1981), polyglutamate methotrexate derivatives (Galivan, MoI. Pharmacol. 77(1): 105- 10, 1980), halogentated methotrexate derivatives (Fox, JNCI 58(4):J955-8, 1977), 8-alkyl-7,8-dihydro analogues (Chaykovsky et al., J. Med. Chem. 20(10):J1323-7, 1977), 7-methyl methotrexate derivatives and dichloromethotrexate (Rosowsky & Chen, J. Med. Chem. 77(12):J1308-11 , 1974), lipophilic methotrexate derivatives and 3',5'-dichloromethotrexate (Rosowsky, J. Med. Chem. 76(10):J1190-3, 1973), deaza amethopterin analogues (Montgomery et al., Ann. N. Y. Acad. Sci. 786:J227-34, 1971), MX068 (Pharma Japan, 1658:18, 1999) and cysteic acid and homocysteic acid methotrexate analogues (EPA 0142220);
These compounds are believed to act as antimetabolites of folic acid.
(D) Podophyllotoxins
In another aspect, the therapeutic agent is a podophyllotoxin, or a derivative or an analogue thereof. Exemplary compounds of this type are etoposide or teniposide, which have the following structures:
Figure imgf000149_0001
Other representative examples of podophyllotoxins include Cu(II)- VP-16 (etoposide) complex (Tawa et al., Bioorg. Med. Chem. 6(7):1003-1008, 1998), pyrrolecarboxamidino-bearing etoposide analogues (Ji et al., Bioorg. Med. Chem. Lett. 7(5):607-612, 1997), 4β-amino etoposide analogues (Hu, University of North Carolina Dissertation, 1992), γ-lactone ring-modified arylamino etoposide analogues (Zhou et al., J. Med. Chem. 37(2):287-92, 1994), N-glucosyl etoposide analogue (Allevi et al., Tetrahedron Lett. 34(45):7313-16, 1993), etoposide A-ring analogues (Kadow et al., Bioorg. Med. Chem. Lett. 2(1): 17-22, 1992), 4'-deshydroxy-4'-methyl etoposide (Saulnier et al., Bioorg. Med. Chem. Lett. 2(10): 1213-18, 1992), pendulum ring etoposide analogues (Sinha et al., Eur. J. Cancer 26(5): 590-3, 1990) and E-ring desoxy etoposide analogues (Saulnier et al., J. Med. Chem. 32(7):1418-20, 1989).
These compounds are believed to act as topoisomerase Il inhibitors and/or DNA cleaving agents.
(E) Camptothecins
In another aspect, the therapeutic agent is camptothecin, or an analogue or derivative thereof. Camptothecins have the following general structure.
Figure imgf000149_0002
In this structure, X is typically O, but can be other groups, e.g., NH in the case of 21-Iactam derivatives. Ri is typically H or OH, but may be other groups, e.g., a terminally hydroxylated Ci_3 alkane. R2 is typically H or an amino containing group such as (CH3)2NHCH2, but may be other groups e.g., NO2, NH2, halogen (as disclosed in, e.g., U.S. Patent 5,552,156) or a short alkane containing these groups. R3 is typically H or a short alkyl such as C2H5. R4 is typically H but may be other groups, e.g., a methylenedioxy group with Ri.
Exemplary camptothecin compounds include topotecan, irinotecan (CPT-11), 9-aminocamptothecin, 21-lactam-20(S)-camptothecin, lO.H-methylenedioxycamptothecin, SN-38, 9-nitrocamptothecin, 10- hydroxycamptothecin. Exemplary compounds have the structures:
Figure imgf000150_0001
R1 R2 R3
Camptothecin : H H H
Topotecan: OH (CH; ,)2NHCH2 H
SN-38: OH H C2H5
X: O for most analogs, NH for 21 -lactam analogs
Camptothecins have the five rings shown here. The ring labeled E must be intact (the lactone rather than carboxylate form) for maximum activity and minimum toxicity.
Camptothecins are believed to function as topoisomerase I inhibitors and/or DNA cleavage agents.
(F) Hydroxyureas
The therapeutic agent of the present invention may be a hydroxyurea. Hydroxyureas have the following general structure:
Figure imgf000151_0001
Suitable hydroxyureas are disclosed in, for example, U.S. Patent No. 6,080,874, wherein R1 is:
Figure imgf000151_0002
and R2 is an alkyl group having 1-4 carbons and R3 is one of H, acyl, methyl, ethyl, and mixtures thereof, such as a methylether.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,665,768, wherein Ri is a cycloalkenyl group, for example N-[3-[5-(4- fluorophenylthio)-furyl]-2-cyclopenten-1-yl]N-hydroxyurea; R2 is H or an alkyl group having 1 to 4 carbons and R3 is H; X is H or a cation.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 4,299,778, wherein Ri is a phenyl group substituted with one or more fluorine atoms; R2 is a cyclopropyl group; and R3 and X is H.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,066,658, wherein R2 and R3 together with the adjacent nitrogen form:
Figure imgf000151_0003
wherein m is 1 or 2, n is 0-2 and Y is an alkyl group.
In one aspect, the hydroxyurea has the structure:
Figure imgf000151_0004
Hydroxyurea These compounds are thought to function by inhibiting DNA synthesis.
(G) Platinum complexes
In another aspect, the therapeutic agent is a platinum compound. In general, suitable platinum complexes may be of Pt(II) or Pt(IV) and have this basic structure:
Figure imgf000152_0001
wherein X and Y are anionic leaving groups such as sulfate, phosphate, carboxylate, and halogen; R1 and R2 are alkyl, amine, amino alkyl any may be further substituted, and are basically inert or bridging groups. For Pt(II) complexes Z1 and Z2 are non-existent. For Pt(IV) Z1 and Z2 may be anionic groups such as halogen, hydroxy, carboxylate, ester, sulfate or phosphate. See, e.g., U.S. Patent Nos. 4,588,831 and 4,250,189.
Suitable platinum complexes may contain multiple Pt atoms. See, e.g., U.S. Patent Nos. 5,409,915 and 5,380,897. For example bisplatinum and triplatinum complexes of the type:
Figure imgf000153_0001
Exemplary platinum compounds are cisplatin, carboplatin, oxaliplatin, and miboplatin having the structures:
Figure imgf000153_0002
Cisplatin Carboplatin
Figure imgf000153_0003
Oxaliplatin
Figure imgf000153_0004
Other representative platinum compounds include (CPA)2Pt[DOLYM] and (DACH)Pt[DOLYM] cisplatin (Choi et al., Arch. Pharmacal Res. 22(2): 151 -156, 1999), Cis-[PtCI2(4,7-H-5-methyl-7- oxo]1 ,2,4[triazolo[1,5-a]pyrimidine)2] (Navarro et al., J. Med. Chem. 41 (3):332- 338, 1998), [Pt(cis-1 ,4-DACH)(trans-CI2)(CBDCA)] • 1/2MeOH cisplatin (Shamsuddin et al., Inorg. Chem. 36(25):5969-5971 , 1997), 4-pyridoxate diammine hydroxy platinum (Tokunaga et al,, Pharm. Sci. 3(7):353-356, 1997), Pt(II) ... Pt(II) (Pt2[NHCHN(C(CH2)(CH3))]4) (Navarro et al., Inorg. Chem. 35(26):7829-7835, 1996), 254-S cisplatin analogue (Koga et al., Neurol. Res. -/8(3): 244-247, 1996), o-phenylenediamine ligand bearing cisplatin analogues (Koeckerbauer & Bednarski, J. Inorg. Biochem. 62(4):281-298, 1996), trans, cis-[Pt(OAc)2l2(en)] (Kratochwil et al., J. Med. Chem. 39(13):2499-2507, 1996), estrogenic 1 ,2-diarylethylenediamine ligand (with sulfur-containing amino acids and glutathione) bearing cisplatin analogues (Bednarski, J. Inorg. Biochem. 62(1 ):75, 1996), cis-1 ,4-diaminocyclohexane cisplatin analogues (Shamsuddin et al., J. Inorg. Biochem. 67(4):291-301 , 1996), 5' orientational isomer of cis- [Pt(NH3)(4-aminoTEMP-O){d(GpG)}] (Dunham & Lippard, J. Am. Chem. Soc. 117(43): 10702-12, 1995), chelating diamine-bearing cisplatin analogues (Koeckerbauer & Bednarski, J. Pharm. Sci. 84(7):819-23, 1995), 1 ,2- diarylethyleneamine ligand-bearing cisplatin analogues (Otto et al, J. Cancer Res. Clin. Oncol. 727(1):31-8, 1995), (ethylenediamine)platinum(ll) complexes (Pasini et al., J. Chem. Soc, Dalton Trans. 4:579-85, 1995), CI-973 cisplatin analogue (Yang et al., Int. J. Oncol. 5(3):597-602, 1994), cis- diaminedichloroplatinum(ll) and its analogues cis-1 ,1- cyclobutanedicarbosylato(2R)-2-methyl-1 ,4-butanediamineplatinum(ll) and cis- diammine(glycolato)platinum (Claycamp & Zimbrick, J. Inorg. Biochem. 26(4):257-67, 1986; Fan et al., Cancer Res. 48(11):3135-9, 1988; Heiger- Bernays et al., Biochemistry 29(36):8461-6, 1990; Kikkawa et al., J. Exp. Clin. Cancer Res. 72(4):233-40, 1993; Murray et al., Biochemistry 31 (47)Λ 1812-17, 1992; Takahashi et al., Cancer Chemother. Pharmacol. 33(1):31-5, 1993), cis- amine-cyclohexylamine-dichloroplatinum(ll) (Yoshida et al., Biochem. Pharmacol. 48(4):793-9, 1994), gem-diphosphonate cisplatin analogues (FR 2683529), (meso-1 ,2-bis(2,6-dichloro-4-hydroxyplenyl)ethylenediamine) dichloroplatinum(ll) (Bednarski et al., J. Med. Chem. 35(23):4479-85, 1992), cisplatin analogues containing a tethered dansyl group (Hartwig et al., J. Am. Chem. Soc. 774(21):8292-3, 1992), platinum(ll) polyamines (Siegmann et al., Inorg. Met-Containing Polym. Mater., (Proc. Am. Chem. Soc. Int. Symp.), 335- 61 , 1990), cis-(3H)dichloro(ethylenediamine)platinum(ll) (Eastman, Anal. Biochem. 797(2):311-15, 1991), trans-diamminedichloroplatinum(ll) and cis- (Pt(NH3)2(N3-cytosine)CI) (Bellon & Lippard, Biophys. Chem. 35(2-3): 179-88, 1990), 3H-cis-1 ,2-diaminocyclohexanedichloroplatinum(ll) and 3H-cis-1 ,2- diaminocyclohexanemalonatoplatinum (II) (Oswald et al., Res. Commun. Chem. Pathol. Pharmacol. 64(1):41-58, 1989), diaminocarboxylatoplatinum (EPA 296321), trans-(D,1)-1 ,2-diaminocyclohexane carrier ligand-bearing platinum analogues (Wyrick & Chaney, J. Labelled Compd. Radiopharm. 25(4):349-57, 1988), aminoalkylaminoanthraquinone-derived cisplatin analogues (Kitov et al., Eur. J. Med. Chem. 23(4):381-3, 1988), spiroplatin, carboplatin, iproplatin and JM40 platinum analogues (Schroyen et al., Eur. J. Cancer Clin. Oncol. 24(8): 1309-12, 1988), bidentate tertiary diamine-containing cisplatinum derivatives (Orbell et al., Inorg. Chim. Acta 752(2): 125-34, 1988), platinum(ll), platinum(IV) (Liu & Wang, Shandong Yike Daxue Xuebao 24(1 ):35-41 , 1986), cis-diammine(1 ,1-cyclobutanedicarboxylato-)platinum(ll) (carboplatin, JM8) and ethylenediammine-malonatoplatinum(ll) (JM40) (Begg et al., Radiother. Oncol. 9(2): 157-65, 1987), JM8 and JM9 cisplatin analogues (Harstrick et al., Int. J. Androl. 70(1); 139-45, 1987), (NPr4)2((PtCL4).cis-(PtCI2-(NH2Me)2)) (Brammer et al., J. Chem. Soc, Chem. Commun. 6:443-5, 1987), aliphatic tricarboxylic acid platinum complexes (EPA 185225), and cis-dichloro(amino acid)(tert-butylamine)platinum(ll) complexes (Pasini & Bersanetti, Inorg. Chim. Acta 707(4):259-67, 1985). These compounds are thought to function by binding to DNA, i.e., acting as alkylating agents of DNA. As medical implants are made in a variety of configurations and sizes, the exact dose administered may vary with device size, surface area, design and portions of the implant coated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Regardless of the method of application of the drug to the cardiac implant, the preferred anticancer agents, used alone or in combination, may be administered under the following dosing guidelines:
(a) Anthracvclines. Utilizing the anthracycline doxorubicin as an example, whether applied as a polymer coating, incorporated into the polymers which make up the implant components, or applied without a carrier polymer, the total dose of doxorubicin applied to the implant should not exceed 25 mg (range of 0.1 μg to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 μg to 5 mg. The dose per unit area (Ae., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 μg - 100 μg per mm2 of surface area. In a particularly preferred embodiment, doxorubicin should be applied to the implant surface at a dose of 0.1 μg/mm2 - 10 μg/mm2. As different polymer and non-polymer coatings may release doxorubicin at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10"8- 10"4 M of doxorubicin is maintained on the surface. It is necessary to insure that surface drug concentrations exceed concentrations of doxorubicin known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10"4 M; although for some embodiments lower concentrations are sufficient). In a preferred embodiment, doxorubicin is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of doxorubicin (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as doxorubicin is administered at half the above parameters, a compound half as potent as doxorubicin is administered at twice the above parameters, etc.).
Utilizing mitoxantrone as another example of an anthracycline, whether applied as a polymer coating, incorporated into the polymers which make up the implant, or applied without a carrier polymer, the total dose of mitoxantrone applied should not exceed 5 mg (range of 0.01 μg to 5 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 0.1 μg to 3 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 μg - 20 μg per mm2 of surface area. In a particularly preferred embodiment, mitoxantrone should be applied to the implant surface at a dose of 0.05 μg/mm2 - 5 μg/mm2. As different polymer and non-polymer coatings will release mitoxantrone at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10"4- 10"8 M of mitoxantrone is maintained. It is necessary to insure that drug concentrations on the implant surface exceed concentrations of mitoxantrone known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10"5 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, mitoxantrone is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of mitoxantrone (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as mitoxantrone is administered at half the above parameters, a compound half as potent as mitoxantrone is administered at twice the above parameters, etc.). (b) Fluoropyrimidines. Utilizing the fluoropyrimidine 5-fluorouracil as an example, whether applied as a polymer coating, incorporated into the polymers which make up the implant, or applied without a carrier polymer, the total dose of 5-fluorouracil applied should not exceed 250 mg (range of 1.0 μg to 250 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 10 μg to 25 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.05 μg - 200 μg per mm2 of surface area. In a particularly preferred embodiment, 5-fluorouracil should be applied to the implant surface at a dose of 0.5 μg/mm2 - 50 μg/mm2. As different polymer and non-polymer coatings will release 5-fluorouracil at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10"4- 10"7 M of 5-fluorouracil is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of 5-fluorouracil known to be lethal to numerous species of bacteria and fungi (i.e., are in excess of 10"4 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, 5-fluorouracil is released from the implant surface such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of 5- fluorouracil (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as 5- fluorouracil is administered at half the above parameters, a compound half as potent as 5-fluorouracil is administered at twice the above parameters, etc.).. (c) Podophylotoxins. Utilizing the podophylotoxin etoposide as an example, whether applied as a polymer coating, incorporated into the polymers which make up the cardiac implant, or applied without a carrier polymer, the total dose of etoposide applied should not exceed 25 mg (range of 0.1 μg to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 μg to 5 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 μg - 100 μg per mm2 of surface area. In a particularly preferred embodiment, etoposide should be applied to the implant surface at a dose of 0.1 μg/mm2 - 10 μg/mm2. As different polymer and non-polymer coatings will release etoposide at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a concentration of 10"4- 10'7 M of etoposide is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of etoposide known to be lethal to a variety of bacteria and fungi (i.e., are in excess of 10"s M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, etoposide is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of etoposide (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as etoposide is administered at half the above parameters, a compound half as potent as etoposide is administered at twice the above parameters, etc.).
It may be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate and/or podophylotoxins (e.g., etoposide) can be utilized to enhance the antibacterial activity of the composition.
In another aspect, an anti-infective agent (e.g., anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate and/or podophylotoxins (e.g., etoposide)) can be combined with traditional antibiotic and/or antifungal agents to enhance efficacy. The anti-infective agent may be further combined with anti-thrombotic and/or antiplatelet agents (for example, heparin, dextran sulphate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, aspirin, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, ticlopidine, clopidogrel, abcixamab, eptifibatide, tirofiban, streptokinase, and/or tissue plasminogen activator) to enhance efficacy.
In addition to incorporation of the above-mentioned therapeutic agents (i.e., anti-infective agents or fibrosis-inhibiting agents), one or more other pharmaceutically active agents can be incorporated into the present compositions and devices to improve or enhance efficacy. Representative examples of additional therapeutically active agents include, by way of example and not limitation, anti-thrombotic agents, anti-proliferative agents, antiinflammatory agents, neoplastic agents, enzymes, receptor antagonists or agonists, hormones, antibiotics, antimicrobial agents, antibodies, cytokine inhibitors, IMPDH (inosine monophosplate dehydrogenase) inhibitors tyrosine kinase inhibitors, MMP inhibitors, p38 MAP kinase inhibitors, immunosuppressants, apoptosis antagonists, caspase inhibitors, and JNK inhibitors. Implantable implantable pump and sensor devices and compositions for use with implantable pump and sensor devices may further include an anti-thrombotic agent and/or antiplatelet agent and/or a thrombolytic agent, which reduces the likelihood of thrombotic events upon implantation of a medical implant. Within various embodiments of the invention, a device is coated on one aspect with a composition which inhibits fibrosis (and/or restenosis), as well as being coated with a composition or compound which prevents thrombosis on another aspect of the device. Representative examples of anti-thrombotic and/or antiplatelet and/or thrombolytic agents include heparin, heparin fragments, organic salts of heparin, heparin complexes (e.g., benzalkonium heparinate, tridodecylammonium heparinate), dextran, sulfonated carbohydrates such as dextran sulphate, Coumadin, coumarin, heparinoid, danaparoid, argatroban chitosan sulfate, chondroitin sulfate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, acetylsalicylic acid, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, streptokinase, factor Xa inhibitors, such as DX9065a, magnesium, and tissue plasminogen activator. Further examples include plasminogen, lys-plasminogen, alpha-2-antiplasmin, urokinase, aminocaproic acid, ticlopidine, clopidogrel, trapidil (triazolopyrimidine), naftidrofuryl, auriritricarboxylic acid and glycoprotein llb/llla inhibitors such as abcixamab, eptifibatide, and tirogiban. Other agents capable of affecting the rate of clotting include glycosaminoglycans, danaparoid, 4- hydroxycourmarin, warfarin sodium, dicumarol, phenprocoumon, indan-1 ,3- dione, acenocoumarol, anisindione, and rodenticides including bromadiolone, brodifacoum, diphenadione, chlorophacinone, and pidnone.
Compositions for use with implantable pump and sensor devices may be or include a hydrophilic polymer gel that itself has anti-thrombogenic properties. For example, the composition can be in the form of a coating that can comprise a hydrophilic, biodegradable polymer that is physically removed from the surface of the device over time, thus reducing adhesion of platelets to the device surface. The gel composition can include a polymer or a blend of polymers. Representative examples include alginates, chitosan and chitosan sulfate, hyaluronic acid, dextran sulfate, PLURONIC polymers (e.g., F-127 or F87), chain extended PLURONIC polymers, various polyester-polyether block copolymers of various configurations (e.g., AB, ABA, or BAB, where A is a polyester such as PLA, PGA, PLGA, PCL or the like), examples of which include MePEG-PLA, PLA-PEG-PLA, and the like). In one embodiment, the anti-thrombotic composition can include a crosslinked gel formed from a combination of molecules (e.g., PEG) having two or more terminal electrophilic groups and two or more nucleophilic groups.
Implantable pump and sensor devices and compositions for use with implantable pump and sensor devices may further include a compound which acts to have an inhibitory effect on pathological processes in or around the treatment site. In certain aspects, the agent may be selected from one of the following classes of compounds: anti-inflammatory agents (e.g., dexamethasone, cortisone, fludrocortisone, prednisone, prednisolone, 6α- methylprednisolone, triamcinolone, betamethasone, and aspirin); MMP inhibitors (e.g., batimistat, marimistat, TIMP's representative examples of which are included in U.S. Patent Nos. 5,665,777; 5,985,911 ; 6,288,261 ;
5,952,320; 6,441 ,189; 6,235,786; 6,294,573; 6,294,539; 6,563,002; 6,071 ,903;
6,358,980; 5,852,213; 6,124,502; 6,160,132; 6,197,791 ; 6,172,057; 6,288,086;
6,342,508; 6,228,869; 5,977,408; 5,929,097; 6,498,167; 6,534,491 ; 6,548,524; 5,962,481 ; 6,197,795; 6,162,814; 6,441 ,023; 6,444,704; 6,462,073; 6,162,821 ;
6,444,639; 6,262,080; 6,486,193; 6,329,550; 6,544,980; 6,352,976; 5,968,795;
5,789,434; 5,932,763; 6,500,847; 5,925,637; 6,225,314; 5,804,581 ; 5,863,915;
5,859,047; 5,861 ,428; 5,886,043; 6,288,063; 5,939,583; 6,166,082; 5,874,473;
5,886,022; 5,932,577; 5,854,277; 5,886,024; 6,495,565; 6,642,255; 6,495,548; 6,479,502; 5,696,082; 5,700,838; 6,444,639; 6,262,080; 6,486,193; 6,329,550;
6,544,980; 6,352,976; 5,968,795; 5,789,434; 5,932,763; 6,500,847; 5,925,637;
6,225,314; 5,804,581 ; 5,863,915; 5,859,047; 5,861 ,428; 5,886,043; 6,288,063;
5,939,583; 6,166,082; 5,874,473; 5,886,022; 5,932,577; 5,854,277; 5,886,024;
6,495,565; 6,642,255; 6,495,548; 6,479,502; 5,696,082; 5,700,838; 5,861 ,436; 5,691 ,382; 5,763,621 ; 5,866,717; 5,902,791 ; 5,962,529; 6,017,889; 6,022,873;
6,022,898; 6,103,739; 6,127,427; 6,258,851 ; 6,310,084; 6,358,987; 5,872,152;
5,917,090; 6,124,329; 6,329,373; 6,344,457; 5,698,706; 5,872,146; 5,853,623;
6,624,144; 6,462,042; 5,981 ,491 ; 5,955,435; 6,090,840; 6,114,372; 6,566,384;
5,994,293; 6,063,786; 6,469,020; 6,118,001 ; 6,187,924; 6,310,088; 5,994,312; 6,180,611 ; 6,110,896; 6,380,253; 5,455,262; 5,470,834; 6,147,114; 6,333,324;
6,489,324; 6,362,183; 6,372,758; 6,448,250; 6,492,367; 6,380,258; 6,583,299; 5,239,078; 5,892,112; 5,773,438; 5,696,147; 6,066,662; 6,600,057; 5,990,158;
5,731 ,293; 6,277,876; 6,521 ,606; 6,168,807; 6,506,414; 6,620,813; 5,684,152;
6,451 ,791 ; 6,476,027; 6,013,649; 6,503,892; 6,420,427; 6,300,514; 6,403,644;
6,177,466; 6,569,899; 5,594,006; 6,417,229; 5,861 ,510; 6,156,798; 6,387,931 ; 6,350,907; 6,090,852; 6,458,822; 6,509,337; 6,147,061 ; 6,114,568; 6,118,016;
5,804,593; 5,847,153; 5,859,061 ; 6,194,451 ; 6,482,827; 6,638,952; 5,677,282;
6,365,630; 6,130,254; 6,455,569; 6,057,369; 6,576,628; 6,110,924; 6,472,396;
6,548,667; 5,618,844; 6,495,578; 6,627,411 ; 5,514,716; 5,256,657; 5,773,428;
6,037,472; 6,579,890; 5,932,595; 6,013,792; 6,420,415; 5,532,265; 5,639,746; 5,672,598; 5,830,915; 6,630,516; 5,324,634; 6,277,061 ; 6,140,099; 6,455,570;
5,595,885; 6,093,398; 6,379,667; 5,641 ,636; 5,698,404; 6,448,058; 6,008,220;
6,265,432; 6,169,103; 6,133,304; 6,541 ,521 ; 6,624,196; 6,307,089; 6,239,288;
5,756,545; 6,020,366; 6,117,869; 6,294,674; 6,037,361 ; 6,399,612; 6,495,568;
6,624,177; 5,948,780; 6,620,835; 6,284,513; 5,977,141 ; 6,153,612; 6,297,247; 6,559,142; 6,555,535; 6,350,885; 5,627,206; 5,665,764; 5,958,972; 6,420,408;
6,492,422; 6,340,709; 6,022,948; 6,274,703; 6,294,694; 6,531 ,499; 6,465,508;
6,437,177; 6,376,665; 5,268,384; 5,183,900; 5,189,178; 6,511 ,993; 6,617,354;
6,331 ,563; 5,962,466; 5,861 ,427; 5,830,869; and 6,087,359), cytokine inhibitors
(chlorpromazine, mycophenolic acid, rapamycin, 1α-hydroxy vitamin D3), IMPDH (inosine monophosplate dehydrogenase) inhibitors (e.g., mycophenolic acid, ribaviran, aminothiadiazole, thiophenfurin, tiazofurin, viramidine)
(Representative examples are included in U.S. Patent, Nos. 5,536,747;
5,807,876; 5,932,600; 6,054,472; 6,128,582; , 6,344,465; 6,395,763; 6,399,773;
6,420,403; 6,479,628; 6,498,178; 6,514,979; 6,518,291; 6,541,496; 6,596,747; 6,617,323; and 6,624,184, U.S. PatentApplication Nos.2002/0040022A1,
2002/0052513A1, 2002/0055483A1, 2002/0068346A1, 2002/0111378A1,
2002/0111495A1, 2002/0123520A1, 2002/0143176A1, 2002/0147160A1,
2002/0161038A1, 2002/0173491A1, 2002/0183315A1, 2002/0193612A1,
2003/0027845A1, 2003/0068302A1, 2003/0105073A1, 2003/0130254A1, 2003/0143197A1, 2003/0144300A1, 2003/0166201A1, 2003/0181497A1,
2003/0186974A1, 2003/0186989A1, and 2003/0195202A1, and PCT Publication Nos. WO 00/24725A1 , WO 00/25780A1 , WO 00/26197A1 , WO 00/51615A1 , WO 00/56331A1, WO 00/73288A1 , WO 01/00622A1, WO 01/66706A1 , WO 01/79246A2, WO 01/81340A2, WO 01/85952A2, WO 02/16382A1 , WO 02/18369A2, WO 02/051814A1, WO 02/057287A2, WO 02/057425A2, WO 02/060875A1 , WO 02/060896A1 , WO 02/060898A1 , WO 02/068058A2, WO 03/020298A1, WO 03/037349A1, WO 03/039548A1, WO 03/045901A2, WO 03/047512A2, WO 03/053958A1 , WO 03/055447A2, WO 03/059269A2, WO 03/063573A2, WO 03/087071 A1, WO 99/001545A1 , WO 97/40028A1, WO 97/41211A1 , WO 98/40381A1, and WO 99/55663A1), p38 MAP kinase inhibitors (MAPK) (e.g., GW-2286, CGP-52411 , BIRB-798, SB220025, RO-320-1195, RWJ-67657, RWJ-68354, SCIO-469) (Representative examples are included in U.S. Patent Nos. 6,300,347; 6,316,464; 6,316,466; 6,376,527; 6,444,696; 6,479,507; 6,509,361 ; 6,579,874, and 6,630,485, and U.S. Patent Application Publication Nos. 2001/0044538A1 , 2002/0013354A1 , 2002/0049220A1, 2002/0103245A1, 2002/0151491 A1, 2002/0156114A1 , 2003/0018051A1 , 2003/0073832A1 , 2003/0130257A1 , 2003/0130273A1 , 2003/0130319A1, 2003/0139388A1, 2003/0139462A1, 2003/0149031 A1 , 2003/0166647A1, and 2003/0181411A1 , and PCT Publication Nos. WO 00/63204A2, WO 01/21591 A1 , WO 01/35959A1 , WO 01/74811A2, WO 02/18379A2, WO 02/064594A2, WO 02/083622A2, WO 02/094842A2, WO 02/096426A1 , WO 02/101015A2, WO 02/103000A2, WO 03/008413A1, WO 03/016248A2, WO 03/020715A1 , WO 03/024899A2, WO 03/031431 A1 , WO 03/040103A1 , WO 03/053940A1 , WO 03/053941 A2, WO 03/063799A2, WO 03/079986A2, WO 03/080024A2, WO 03/082287A1 , WO 97/44467A1 , WO 99/01449A1 , and WO 99/58523A1), and immunomodulatory agents (rapamycin, everolimus, ABT-578, azathioprine azithromycin, analogues of rapamycin, including tacrolimus and derivatives thereof (e.g., EP 0184162B1 and those described in U.S. Patent No. 6,258,823) and everolimus and derivatives thereof (e.g., U.S. Patent No. 5,665,772). Further representative examples of sirolimus analogues and derivatives include ABT-578 and those found in PCT Publication Nos. WO 97/10502, WO 96/41807, WO 96/35423, WO 96/03430, WO 96/00282, WO 95/16691 , WO 95/15328, WO 95/07468, WO 95/04738, WO 95/04060, WO 94/25022, WO 94/21644, WO 94/18207, WO 94/10843, WO 94/09010, WO 94/04540, WO 94/02485, WO 94/02137, WO 94/02136, WO 93/25533, WO 93/18043, WO 93/13663, WO 93/11130, WO 93/10122, WO 93/04680, WO 92/14737, and WO 92/05179 and in U.S. Patent Nos. 6,342,507; 5,985,890; 5,604,234; 5,597,715; 5,583,139; 5,563,172; 5,561 ,228; 5,561 ,137; 5,541 ,193; 5,541 ,189; 5,534,632; 5,527,907; 5,484,799; 5,457,194; 5,457,182; 5,362,735; 5,324,644; 5,318,895; 5,310,903; 5,310,901 ; 5,258,389; 5,252,732; 5,247,076; 5,225,403; 5,221 ,625; 5,210,030; 5,208,241 ; 5,200,411 ; 5,198,421 ; 5,147,877; 5,140,018; 5,116,756; 5,109,112; 5,093,338; and 5,091 ,389.
Other examples of biologically active agents which may be combined with implantable pump and sensor devices according to the invention include tyrosine kinase inhibitors, such as imantinib, ZK-222584, CGP-52411 , CGP-53716, NVP-AAK980-NX, CP-127374, CP-564959, PD-171026, PD- 173956, PD-180970, SU-0879, and SKI-606; MMP inhibitors such as nimesulide, PKF-241-466, PKF-242-484, CGS-27023A, SAR-943, primomastat, SC-77964, PNU-171829, AG-3433, PNU-142769, SU-5402, and dexlipotam; p38 MAP kinase inhibitors such as include CGH-2466 and PD-98-59; immunosuppressants such as argyrin B, macrocyclic lactone, ADZ-62-826,
CCI-779, tilomisole, amcinonide, FK-778, AVE-1726, and MDL-28842; cytokine inhibitors such as TNF-484A, PD-172084, CP-293121 , CP-353164, and PD- 168787; NFKB inhibitors, such as, AVE-0547, AVE-0545, and IPL-576092; HMGCoA reductase inhibitors, such as, pravestatin, atorvastatin, fluvastatin, dalvastatin, glenvastatin, pitavastatin, CP-83101 , U-20685; apoptosis antagonist (e.g., troloxamine, TCH-346 (N-methyl-N-propargyl-10-aminomethyl- dibenzo(b,f)oxepin); and caspase inhibitors (e.g., PF-5901 (benzenemethanol, alpha-pentyl-3-(2-quinolinylmethoxy)-), and JNK inhibitor (e.g., AS-602801).
In another aspect, the implantable pump and sensor devices may further include an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
In certain aspects, a polymeric composition comprising a fibrosis- inhibiting agent is combined with an agent that can modify metabolism of the agent in vivo to enhance efficacy of the fibrosis-inhibiting agent. One class of therapeutic agents that can be used to alter drug metabolism includes agents capable of inhibiting oxidation of the anti-scarring agent by cytochrome P450 (CYP). In one embodiment, compositions are provided that include a fibrosis- inhibiting agent (e.g., The following agents are exemplary compounds: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin) and a CYP inhibitor, which may be combined (e.g., coated) with any of the devices described herein. Representative examples of CYP inhibitors include flavones, azole antifungals, macrolide antibiotics, HIV protease inhibitors, and anti-sense oligomers. Devices comprising a combination of a fibrosis-inhibiting agent and a CYP inhibitor may be used to treat a variety of proliferative conditions that can lead to undesired scarring of tissue, including intimal hyperplasia, surgical adhesions, and tumor growth. Within various embodiments of the invention, a device incorporates or is coated on one aspect, portion or surface, portion or surface with a composition which inhibits fibrosis (and/or restenosis), as well as with a composition or compound which promotes or stimulates fibrosis on another aspect, portion or surface, portion or surface of the device. Compounds that promote or stimulate fibrosis can be identified by, for example, the in vivo (animal) models provided in Examples 47-50. Representative examples of agents that promote fibrosis include siik and other irritants (e.g., talc, wool (including animal wool, wood wool, and synthetic wool), talcum powder, copper, metallic beryllium (or its oxides), quartz dust, silica, crystalline silicates), polymers (e.g., polylysine, polyurethanes, poly(ethylene terephthalate), PTFE, poly(alkylcyanoacrylates), and poly(ethylene-co-vinylacetate); vinyl chloride and polymers of vinyl chloride; peptides with high lysine content; growth factors and inflammatory cytokines involved in angiogenesis, fibroblast migration, fibroblast proliferation, ECM synthesis and tissue remodeling, such as epidermal growth factor (EGF) family, transforming growth factor-α (TGF- α), transforming growth factor-β (TGF-β-1 , TGF-β-2, TGF-β-3, platelet-derived growth factor (PDGF), fibroblast growth factor (acidic - aFGF; and basic - bFGF), fibroblast stimulating factor-1 , activins, vascular endothelial growth factor (including VEGF-2, VEGF- 3, VEGF-A, VEGF-B, VEGF-C, placental growth factor - PIGF), angiopoietins, insulin-like growth factors (IGF), hepatocyte growth factor (HGF), connective tissue growth factor (CTGF), myeloid colony-stimulating factors (CSFs), monocyte chemotactic protein, granulocyte-macrophage colony-stimulating factors (GM-CSF), granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), erythropoietin, interleukins (particularly IL-1 , IL-8, and IL-6), tumor necrosis factor-α (TNFα), nerve growth factor (NGF), interferon-α, interferon-β, histamine, endothelin-1 , angiotensin II, growth hormone (GH), and synthetic peptides, analogues or derivatives of these factors are also suitable for release from specific implants and devices to be described later. Other examples include CTGF (connective tissue growth factor); inflammatory microcrystals (e.g., crystalline minerals such as crystalline silicates); bromocriptine, methylsergide, methotrexate, chitosan, N-carboxybutyl chitosan, carbon tetrachloride, thioacetamide, fibrosin, ethanol, bleomycin, naturally occurring or synthetic peptides containing the Arg-Gly-Asp (RGD) sequence, generally at one or both termini (see, e.g., U.S. Patent No. 5,997,895), and tissue adhesives, such as cyanoacrylate and crosslinked poly(ethylene glycol) - methylated collagen compositions. Other examples of fibrosis-inducing agents include bone morphogenic proteins (e.g., BMP-2, BMP- 3, BMP-4, BMP-5, BMP-6 (VgM), BMP-7 (OP-1), BMP-8, BMP-9, BMP-10, BMP-11 , BMP-12, BMP-13, BMP-14, BMP-15, and BMP-16. Of these, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, and BMP-7 are of particular utility. Bone morphogenic proteins are described, for example, in U.S. Patent Nos. 4,877,864; 5,013,649; 5,661 ,007; 5,688,678; 6,177,406; 6,432,919; and 6,534,268 and Wozney, J. M., et al. (1988) Science: 242(4885); 1528-1534.
Other representative examples of fibrosis-inducing agents include components of extracellular matrix (e.g., fibronectin, fibrin, fibrinogen, collagen (e.g., bovine collagen), including fibrillar and non-fibrillar collagen, adhesive glycoproteins, proteoglycans (e.g., heparin sulfate, chondroitin sulfate, dermatan sulfate), hyaluronan, secreted protein acidic and rich in cysteine (SPARC), thrombospondins, tenacin, and cell adhesion molecules (including integrins, vitronectin, fibronectin, laminin, hyaluronic acid, elastin, bitronectin), proteins found in basement membranes, and fibrosin) and inhibitors of matrix metalloproteinases, such as TIMPs (tissue inhibitors of matrix metalloproteinases) and synthetic TIMPs, such as, e.g., marimistat, batimistat, doxycycline, tetracycline, minocycline, TROCADE, Ro-1130830, CGS 27023A, and BMS-275291 and analogues and derivatives thereof.
Although the above therapeutic agents have been provided for the purposes of illustration, it may be understood that the present invention is not so limited. For example, although agents are specifically referred to above, the present invention may be understood to include analogues, derivatives and conjugates of such agents. As an illustration, combretastatin A4may be understood to refer to not only the common chemically available form of combretastatin, but analogues (e.g., combretastatin A2, A3, A5, A6, as noted above) and combretastatin conjugates. In addition, as will be evident to one of skill in the art, although the agents set forth above may be noted within the context of one class, many of the agents listed in fact have multiple biological activities. Further, more than one therapeutic agent may be utilized at a time (i.e., in combination), or delivered sequentially. Dosages
Since implantable sensor and implantable pumps (and their drug delivery catheters or ports) are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, as described above, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose (i.e., amount) per unit area of the portion of the device being coated. Surface area can be measured or determined by methods known to one of ordinary skill in the art. Total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single systemic dose application. In certain embodiments, the drug is released in effective concentrations for a period ranging from 1 - 90 days. Regardless of the method of application of the drug to the device, the fibrosis-inhibiting agents, used alone or in combination, may be administered under the following dosing guidelines:
As described above, implantable sensors and pumps may be used in combination with a composition that includes an anti-scarring agent. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg- 1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
For high potency drugs (approximately 1-10OnM) IC50 range in assays described herein), the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10'8- 10'4 M of agent should be maintained on the implant or barrier surface. For mid-potency agents (approximately 100-500 nM IC50 range), the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2. For low potency drugs (approximately 500-1000 nM IC50 range), the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2. It may be apparent to one of skill in the art that potentially any anti- fibrosis agent described above may be utilized alone, or in combination, in the practice of this embodiment. In various aspects, the present invention provides an implantable sensor or pump that contains an anti-fibrosing agent listed below in a dosage as set forth above: 1) an anti-fibrotic agent that inhibits cell regeneration, 2) an anti-fibrotic agent that inhibits angiogenesis, 3) an anti-fibrotic agent that inhibits fibroblast migration, 4) an anti-fibrotic agent that inhibits fibroblast proliferation, 5) an anti-fibrotic agent that inhibits deposition of extracellular matrix, 6) an anti- fibrotic agent inhibits tissue remodeling, 7) an adensosine A2A receptor antagonist, 8) an AKT inhibitor, 9) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA), 10) an alpha 4 integrin antagonist, 11) an alpha 7 nicotinic receptor agonist, 12) an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005
(GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof, 13) an apoptosis antagonist, 14) an apoptosis activator, 15) a beta 1 integrin antagonist, 16) a beta tubulin inhibitor, 17) a blocker of enzyme production in Hepatitis C, 18) a Bruton's tyrosine kinase inhibitor, 19) a calcineurin inhibitor, 20) a caspase 3 inhibitor, 21) a CC chemokine receptor antagonist, 22) a cell cycle inhibitor selected from the group consisting of SNS- 595 (Sunesis), synthadotin, KRX-0403, homoharringtonine, and an analogue or derivative thereof, 23) a cathepsin B inhibitor, 24) a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof, 25) a cathepsin L inhibitor, 26) a CD40 antagonist, 27) a chemokine receptor agonist, 28) a chymase inhibitor, 29) a collagenase antagonist, 30) a CXCR antagonist, 31) a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof, 32) a cyclooxygenase 1 inhibitor, 33) a DHFR inhibitor, 34) a dual integrin inhibitor, 35) an elastase inhibitor, 36) an elongation factor-1 alpha inhibitor, 37) an endothelial growth factor antagonist, 38) an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No. 125697-92-9), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof, 39) an endotoxin antagonist, 40) an epothilone and tubulin binder, 41) an estrogen receptor antagonist, 42) an FGF inhibitor, 43) a farnexyl transferase inhibitor, 44) a famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof, 45) an FLT-3 kinase inhibitor, 46a) an FGF receptor kinase inhibitor, 47) a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, and an analogue or derivative thereof, 48) a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17- dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1l,4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No. 12772-57-5), and an analogue or derivative thereof, 49) a histone deacetylase inhibitor, 50) an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No. 143201-11-0), and an analogue or derivative thereof, 51) an ICAM inhibitor, 52) an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma- Lerads), and an analogue or derivative thereof, 53) an IL-2 inhibitor, 54) an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof, 55) an IMPDH (inosine monophosphate), 56) an integrin antagonist, 57) an interleukin antagonist, 58) an inhibitor of type III receptor tyrosine kinase, 59) an irreversible inhibitor of enzyme methionine aminopeptidase type 2, 60) an isozyme selective delta protein kinase C inhibitor, 61) a JAK3 enzyme inhibitor, 62) a JNK inhibitor, 63) a kinase inhibitor, 64) a kinesin antagonist, 65) a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605
(CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85- 2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof, 66) a MAP kinase inhibitor, 67) a matrix metalloproteinase inhibitor, 68) an MCP-CCR2 inhibitor, 69) an mTOR inhibitor, 70) an mTOR kinase inhibitor,71) a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gnklf (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, dolastatin 15 (CAS No. 123884-00-4), vincamine, and an analogue or derivative thereof, 72) an MIF inhibitor, 73) an MMP inhibitor, 74) a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476- 64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof, 75) an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), Bay 11-7085, and an analogue or derivative thereof, 76) a nitric oxide agonist, 77) an ornithine decarboxylase inhibitor, 78) a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol- Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74-6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof, 79) a palmitoyl-protein thioesterase inhibitor, 80) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof, 81) a peroxisome proliferators-activated receptor agonist selected from the group consisting of (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof, 82) a phosphatase inhibitor, 83) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66- 5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof, 84) a PKC inhibitor, 85) a platelet activating factor antagonist, 86) a platelet-derived growth factor receptor kinase inhibitor, 87) a prolyl hydroxylase inhibitor, 88) a polymorphonuclear neutrophil inhibitor, 89) a protein kinase B inhibitor, 90) a protein kinase C stimulant, 91) a purine nucleoside analogue, 92) a purinoreceptor P2X antagonist, 93) a Raf kinase inhibitor, 94) a reversible inhibitor of ErbB1 and ErbB2, 95) a ribonucleoside triphosphate reductase inhibitor, 96) an SDF-1 antagonist, 97) a sheddase inhibitor, 98) an SRC inhibitor, 99) a stromelysin inhibitor, 100) an Syk kinase inhibitor, 101) a telomerase inhibitor, 102) a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof, 103) a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57-7/188913- 58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130- 73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y1S Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof, 104) a tumor necrosis factor antagonist, 105) a Toll receptor inhibitor, 106) a tubulin antagonist, 107) a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD- 2171 orAZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), herbimycin A, and an analogue or derivative thereof, 108) a VEGF inhibitor, 109) a vitamin D receptor agonist, 110) ZD-6474 (an angiogenesis inhibitor), 111) AP-23573 (an mTOR inhibitor), 112) synthadotin (a tubulin antagonist), 113) S-0885 (a collagenase inhibitor), 114) aplidine (an elongation factor- 1 alpha inhibitor), 115) ixabepilone (an epithilone), 116) IDN-5390 (an angiogenesis inhibitor and an FGF inhibitor), 117) SB-2723005 (an angiogenesis inhibitor), 118) ABT-518 (an angiogenesis inhibitor), 119) combretastatin (an angiogenesis inhibitor), 120) anecortave acetate (an angiogenesis inhibitor), 121) SB-715992 (a kinesin antagonist), 122) temsirolimus (an mTOR inhibitor), and 123) adalimumab (a TNFα antagonist), 124) erucylphosphocholine (an ATK inhibitor), 125) alphastatin (an angiogenesis inhibitor), 126) bortezomib (an NF Kappa B inhibitor), 127) etanercept (a TNFα antagonist and TACE inhibitor), 128) humicade (a TNFα inhibitor), and 129) gefitinib (a tyrosine kinase inhibitor), 130) a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones), 131) an alpha adrenergic receptor antagonist, 132) an anti-psychotic compound, 133) a CaM kinase Il inhibitor, 134) a G protein agonist, 135) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof, 136) an anti-microbial agent, 137) a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32- 8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof, 138) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof, 139) a D2 dopamine receptor antagonist, 140) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, 141) a dopamine antagonist, an anesthetic compound, 142) a clotting factor, 143) a lysyl hydrolase inhibitor, 144) a muscarinic receptor inhibitor, 145) a superoxide anion generator, 146) a steroid, 147) an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514, spermine tetrahydrochloride, NG-methyl-L- arginine acetate salt, galardin, and an analogue or derivative thereof, 148) a diuretic, 149) an anti-coagulant, 150) a cyclic GMP agonist, 151) an adenylate cyclase agonist, 152) an antioxidant, 153) a nitric oxide synthase inhibitor, 154) an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof, 155) a DNA synthesis inhibitor, 156) a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof, 157) a DNA methylation inhibitor, 158) a NSAID agent, 159) a peptidylglycine alpha- hydroxylating monooxygenase inhibitor, 160) an MEK1/MEK 2 inhibitor, 161) a NO synthase inhibitor, 162) a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof, 163) an ACE inhibitor, 164) a glycosylation inhibitor, 165) an intracellular calcium influx inhibitor, 166) an anti-emetic agent, 167) an acetylcholinesterase inhibitor, 168) an ALK-5 receptor antagonist, 169) a RAR/RXT antagonist, 170) an elF-2a inhibitor, 171) an S-adenosyl-L-homocysteine hydrolase inhibitor, 172) an estrogen agonist, 173) a serotonin receptor inhibitor, 174) an anti-thrombotic agent, 175) a tryptase inhibitor, 176) a pesticide, 177) a bone mineralization promoter, 178) a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, 179) an anti-inflammatory compound, 180) a DNA methylation promoter, 181) an anti-spasmodic agent, 182) a protein synthesis inhibitor, 183) an α-glucosidase inhibitor, 184) a calcium channel blocker, 185) a pyruvate dehydrogenase activator, 186) a prostaglandin inhibitor, 187) a sodium channel inhibitor, 188) a serine protease inhibitor, 189) an intracellular calcium flux inhibitor, 190) a JAK2 inhibitor; 191) an androgen inhibitor, 192) an aromatase inhibitor, 193) an anti-viral agent, 194) a 5-HT inhibitor, 195) an FXR antagonist, 196) an actin polymerization and stabilization promoter, 197) an AXOR12 agonist, 198) an angiotensin Il receptor agonist, 199) a platelet aggregation inhibitor, 200) a CB1/CB2 receptor agonist, 201) a norepinephrine reuptake inhibitor, 202) a selective serotonin reuptake inhibitor, 203) a reducing agent, 204) Isotretinoin, 205) radicicol, 206) clobetasol propionate, 207) homoharringtonine, 208) trichostatin A, 209) brefeldin A, 210) thapsigargin, 211) dolastatin 15, 212) cerivastatin, 213) jasplakinolide, 214) herbimycin A, 215) pirfenidone, 216) vinorelbine, 217) 17-DMAG, 218) tacrolimus, 219) loteprednol etabonate, 220) juglone, 221) prednisolone, 222) puromycin, 223) 3-BAABE, 224) cladribine, 225) mannose-6-phosphate, 226) 5-azacytidine, 227) Ly333531 (ruboxistaurin), 228) simvastatin, and 229) an immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
Provided below are exemplary dosage ranges for a variety of anti- scarring agents which can be used in conjunction with devices in accordance with the invention. (A) Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10' 8- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10' 8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin Antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10'4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10' 8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"s- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF Kappa B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation Factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should to be maintained on the implant or barrier surface. D. Delivery of Therapeutic Agents or Compositions and Generating lmplatnable Sensors or Pumps That Comprise Therapeutic Agents or Compositions
In the practice of this invention, drug-coated or drug-impregnated implants and medical devices are provided which inhibit fibrosis in and around the implantable sensor or implantable pump. Within various embodiments, fibrosis is inhibited by local, regional or systemic release of specific pharmacological agents that become localized to the tissue adjacent to the device or implant. There are numerous implantable sensors or implantable pumps where the occurrence of a f ibrotic reaction will adversely affect the functioning of the device or the biological problem for which the device was implanted or used. Typically, fibrotic encapsulation of the device (or the growth of fibrous tissue between the device and the target tissue) slows, impairs, or interrupts detection (sensors) or drug delivery (pumps) to/from the device to/from the tissue. This can cause the device to function suboptimally or not at all, negatively affect disease management, and/or shorten the lifespan of the device. There are numerous methods available for optimizing delivery of the fibrosis-inhibiting agent to the site of the intervention and several of these are described below.
1. Delivery of Therapeutic Agents via Implantable Sensors or Pumps and Generating Implantable Sensors or Pumps That Comprise Fibrosis-inhibiting Agents
Medical devices or implants of the present invention are coated with, or adapted to release an agent which inhibits fibrosis on the surface of, or around, the implantable sensor and/or implantable pump. In one aspect, the present invention provides implantable sensors and implantable pumps that include an anti-scarring agent or a composition that includes an anti-scarring agent such that the overgrowth of fibrous or granulation tissue is inhibited or reduced. Methods for incorporating fibrosis-inhibiting compositions onto or into implantable sensors and implantable pumps include: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis- inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) by inserting the device into a sleeve or mesh which is comprised of, or coated with, a fibrosis-inhibiting composition, (f) constructing the device itself (or a portion of the device such as the detector, drug delivery catheter or port) with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. Each of these methods illustrates an approach for combining an implantable sensor or an implantable pump with a fibrosis-inhibiting (also referred to herein as anti-scarring) agent according to the present invention.
For these devices, the coating process can be performed in such a manner as to coat all or parts (such as the sensor or the drug delivery catheter/port) of the entire device with the fibrosis-inhibiting composition. In addition to, or alternatively, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the implantable sensor or implantable pump such that the fibrosis-inhibiting agent is incorporated into the final product. In these manners, a medical device may be prepared which has a coating, where the coating is, e.g., uniform, non-uniform, continuous, discontinuous, or patterned.
In another aspect, an implantable sensor or drug delivery/catheter/port device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug (i.e., one or more fibrosis-inhibiting agents). The reservoirs may be formed from divets in the device surface or micropores or channels in the device body. In one aspect, the reservoirs are formed from voids in the structure of the device. The reservoirs may house a single type of drug (e.g., fibrosis-inhibiting agent) or more than one type of drug (e.g., a fibrosis-inhibiting agent and an anti-infective agent). The drug(s) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs. The filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier. In certain embodiments, the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount
(dose) of drug, and each layer may have a different composition to further tailor the amount and type of drug that is released from the substrate. The multi- layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void. Thus, the coating of the medical device may directly contact the implantable device, or it may indirectly contact the device when there is something, e.g., a polymer layer, that is interposed between the device and the coating that contains the fibrosis-inhibiting agent.
In addition to, or as an alternative to incorporating a fibrosis- inhibiting agent onto or into the implantable sensors and implantable pump, the fibrosis-inhibiting agent can be applied directly or indirectly to the tissue adjacent to the implantable sensors and implantable pump (preferably near the interface of the tissue and the detector, drug delivery catheter and/or drug delivery port). This can be accomplished by applying the fibrosis-inhibiting agent, with or without a polymeric, non-polymeric, or secondary carrier: (a) to the device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) prior to, immediately prior to, or during, implantation of the implantable sensors and implantable pump; (c) to the surface of the device and/or the tissue surrounding the implanted pump or sensor (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after implantation; (d) by topical application of the anti-fibrosis agent into the anatomical space where the implantable sensors and implantable pump will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the implantable sensor or implantable pump as a solution, as an infusate, or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies {i.e., combinations of therapeutic agents and combinations with antithrombotic, antiplatelet and/or anti-infective agents) can also be used. In another embodiment, the anti-fibrosing agent can be coated onto the entire device or a portion of the device. In certain embodiments, the agent is present as part of a coating on a surface of the implantable sensor or implantable pump. The coating may partially cover or may completely cover the surface of the implantable sensor or implantable pump. Further, the coating may directly or indirectly contact the implantable sensor or implantable pump. For example, the Implantable sensor or implantable pump may be coated with a first coating and then coated with a second coating that includes the anti- scarring agent.
Implantable sensors and implantable pumps may be coated using a variety of coating methods, including by dipping, spraying, painting, by vacuum deposition, or by any other method known to those of ordinary skill in the art.
As described above, the anti-fibrosing agent can be coated onto the appropriate implantable sensors and implantable pumps using the polymeric coatings described above. In addition to the coating compositions and methods described above, there are various other coating compositions and methods that are known in the art. Representative examples of these coating compositions and methods are described in U.S. Patent. Nos. 6,610,016; 6,358,557; 6,306,176; 6,110,483; 6,106,473; 5,997,517; 5,800,412; 5,525,348; 5,331,027; 5,001,009; 6,562,136; 6,406,754; 6,344,035; 6,254,921; 6,214,901; 6,077,698; 6,603,040; 6,278,018; 6,238,799; 6,096,726, 5,766,158, 5,599,576, 4,119,094; 4,100,309; 6,599,558; 6,369,168; 6,521,283; 6,497,916; 6,251 ,964; 6,225,431; 6,087,462; 6,083,257; 5,739,237; 5,739,236; 5,705,583; 5,648,442; 5,645,883; 5,556,710; 5,496,581; 4,689,386; 6,214,115; 6,090,901; 6,599,448; 6,054,504; 4,987,182; 4,847,324; and 4,642,267; U.S. Patent Application Publication Nos. 2002/0146581 , 2003/0129130, 2001/0026834; 2003/0190420; 2001/0000785; 2003/0059631 ; 2003/0190405; 2002/0146581; 2003/020399; 2001/0026834; 2003/0190420; 2001/0000785; 2003/0059631 ; 2003/0190405; and 2003/020399; and PCT Publication Nos. WO 02/055121; WO 01/57048; WO 01/52915; and WO 01/01957. Within another aspect of the invention, the biologically active fibrosis-inhibiting agent can be delivered with non-polymeric agents. These non-polymeric agents can include sucrose derivatives (e.g., sucrose acetate isobutyrate, sucrose oleate), sterols such as cholesterol, stigmasterol, beta- sitosterol, and estradiol; cholesteryl esters such as cholesteryl stearate; Ci2 - C24 fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, and lignoceric acid; C18 -C36 mono-, di- and triacylglycerides such as glyceryl monooleate, glyceryl monolinoleate, glyceryl monolaurate, glyceryl monodocosanoate, glyceryl monomyristate, glyceryl monodicenoate, glyceryl dipalmitate, glyceryl didocosanoate, glyceryl dimyristate, glyceryl didecenoate, glyceryl tridocosanoate, glyceryl trimyristate, glyceryl tridecenoate, glycerol tristearate and mixtures thereof; sucrose fatty acid esters such as sucrose distearate and sucrose palmitate; sorbitan fatty acid esters such as sorbitan monostearate, sorbitan monopalmitate and sorbitan tristearate; Ci6 -Ci8 fatty alcohols such as cetyl alcohol, myristyl alcohol, stearyl alcohol, and cetostearyl alcohol; esters of fatty alcohols and fatty acids such as cetyl palmitate and cetearyl palmitate; anhydrides of fatty acids such as stearic anhydride; phospholipids including phosphatidylcholine (lecithin), phosphatidylserine, phosphatidylethanolamine, phosphatidylinositol, and lysoderivatives thereof; sphingosine and derivatives thereof; spingomyelins such as stearyl, palmitoyl, and tricosanyl spingomyelins; ceramides such as stearyl and palmitoyl ceramides; glycosphingolipids; lanolin and lanolin alcohols, calcium phosphate, sintered and unscintered hydoxyapatite, zeolites, and combinations and mixtures thereof.
Representative examples of patents relating to non-polymeric delivery systems and their preparation include U.S. Patent Nos. 5,736,152; 5,888,533; 6,120,789; 5,968,542; and 5,747,058.
The fibrosis-inhibiting agent may be delivered as a solution. The fibrosis-inhibiting agent can be incorporated directly into the solution to provide a homogeneous solution or dispersion. In certain embodiments, the solution is an aqueous solution. The aqueous solution may futher include buffer salts, as well as viscosity modifying agents (e.g., hyaluronic acid, alginates, CMC, and the like). In another aspect of the invention, the solution can include a biocompatible solvent, such as ethanol, DMSO, glycerol, PEG-200, PEG-300 or NMP.
Within another aspect of the invention, the fibrosis-inhibiting agent can further comprise a secondary carrier. The secondary carrier can be in the form of microspheres (e.g., PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate), nanospheres (e.g., PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), liposomes, emulsions, microemulsions, micelles (e.g., SDS, block copolymers of the form X-Y, X-Y-X or Y-X-Y where X is a poly(alkylene oxide) or alkyl ether thereof and Y is a polyester (e.g., PLGA, PLLA, PDLLA, PCL polydioxanone)), zeolites or cyclodextrins.
Within another aspect of the invention, these fibrosis-inhibiting agent/secondary carrier compositions can be a) incorporated directly into, or onto, the implantable sensor or implantable pump, b) incorporated into a solution, c) incorporated into a gel or viscous solution, d) incorporated into the composition used for coating the implantable sensor or implantable pump, or e) incorporated into, or onto, the implantable sensor or implantable pump following coating of the implantable sensor or implantable pump with a coating composition. For example, fibrosis-inhibiting agent loaded PLGA microspheres may be incorporated into a polyurethane coating solution which is then coated onto the implantable sensor or implantable pump.
In yet another example, the implantable sensor or implantable pump can be coated with a polyurethane and then allowed to partially dry such that the surface is still tacky. A particulate form of the fibrosis-inhibiting agent or fibrosis-inhibiting agent/secondary carrier can then be applied to all or a portion of the tacky coating after which the device is dried.
In yet another example, the implantable sensor or implantable pump can be coated with one of the coatings described above. A thermal treatment process can then be used to soften the coating, afterwhich the fibrosis-inhibiting agent or the fibrosis-inhibiting agent/secondary carrier is applied to the entire implantable sensor or implantable pump or to a portion of the implantable sensor or implantable pump (e.g., outer surface).
Within another aspect of the invention, the coated Implantable sensor or implantable pump which inhibits or reduces an in vivo fibrotic reaction is further coated with a compound or compositions which delay the release of and/or activity of the fibrosis-inhibiting agent. Representative examples of such agents include biologically inert materials such as gelatin, PLGA/MePEG film, PLA, polyurethanes, silicone rubbers, surfactants, lipids, or polyethylene glycol, as well as biologically active materials such as heparin (e.g., to induce coagulation).
For example, in one embodiment of the invention the fibrosis- inhibiting active agent on the implantable sensor or implantable pump is top- coated with a physical barrier. Such barriers can include non-degradable materials or biodegradable materials such as gelatin, PLGA/MePEG film, PLA, or polyethylene glycol among others. In one embodiment, the rate of diffusion of the therapeutic agent in the barrier coat is slower that the rate of diffusion of the therapeutic agent in the coating layer. In the case of PLGA/ MePEG, once the PLGA/ MePEG becomes exposed to the blood or body fluids, the MePEG will dissolve out of the PLGA, leaving channels through the PLGA to an underlying layer containing the fibrosis-inhibiting agent, which then can then diffuse into the tissue and initiate its biological activity.
In another embodiment of the invention, for example, a particulate form of the active fibrosis-inhibiting agent may be coated onto the implantable sensor or implantable pump using a polymer (e.g., PLG, PLA, polyurethane). A second polymer that dissolves slowly or degrades (e.g., MePEG-PLGA or PLG) and that does not contain the active agent may be coated over the first layer. Once the top layer dissolves or degrades, it exposes the under coating which allows the active agent to be exposed to the treatment site or to be released from the coating. Within another aspect of the invention, the outer layer of the coating of a coated Implantable sensor or implantable pump which inhibits an in vivo fibrotic response is further treated to crosslink the outer layer of the coating. This can be accomplished by subjecting the coated implantable sensor or implantable pump to a plasma treatment process. The degree of crosslinking and nature of the surface modification can be altered by changing the RF power setting, the location with respect to the plasma, the duration of treatment as well as the gas composition introduced into the plasma chamber. Protection of a biologically active surface can also be utilized by coating the implantable sensor or implantable pump surface with an inert molecule that prevents access to the active site through steric hindrance, or by coating the surface with an inactive form of the fibrosis-inhibiting agent, which is later activated. For example, the implantable sensor or implantable pump can be coated with an enzyme, which causes either release of the fibrosis-inhibiting agent or activates the fibrosis-inhibiting agent. Another example of a suitable implantable sensor or implantable pump surface coating includes an anticoagulant such as heparin or heparin quaternary amine complexes (e.g., heparin-benzalkonium chloride complex), which can be coated on top of the fibrosis-inhibiting agent. The presence of the anticoagulant delays coagulation. As the anticoagulant dissolves away, the anticoagulant activity may stop, and the newly exposed fibrosis-inhibiting agent may inhibit or reduce fibrosis from occurring in the adjacent tissue or coating the implantable sensor or implantable pump.
Another example of a suitable implantable sensor or implantable pump surface coating (particularly coatings for drug delivery catheters used in implantable pumps) includes an anti-infective agent such as an antibiotic, 5-FU, mitoxantrone, methotrexate, and/or doxyrubicin which can be incorporated into a coating that may, or may not, also contain a fibrosis-inhibiting agent. The presence of the anti-infective agent prevents infection in the tissues around the implant and can help prevent serious device-related infections (e.g., meningitis with intrathecal drug delivery pumps, peritonitis with intraperitoneal drug delivery pumps, endocarditis with cardiac drug delivery pumps).
In another aspect, the implantable sensor or implantable pump can be coated with an inactive form of the fibrosis-inhibiting agent, which is then activated once the device is deployed. Such activation may be achieved by injecting another material into the treatment area after the implantable sensor or implantable pump (as desribed below) is implanted or after the fibrosis- inhibiting agent has been administered to the treatment area (via injections, spray, wash, drug delivery catheters or balloons). In this aspect, the implantable sensor or implantable pump may be coated with an inactive form of the fibrosis-inhibiting agent. Once the implantable sensor or implantable pump is implanted, the activating substance is injected or applied into, or onto, the treatment site where the inactive form of the fibrosis-inhibiting agent has been applied.
One example of this method includes coating an implantable sensor or implantable pump with a biologically active fibrosis-inhibiting agent, in the usual manner. The coating containing the active fibrosis-inhibiting agent may then be covered with polyethylene glycol and these two substances may then be bonded through an ester bond using a condensation reaction. Prior to the deployment of the implantable sensor or implantable pump, an esterase is injected into the tissue around the outside of the device, which will cleave the bond between the ester and the fibrosis-inhibiting therapeutic, allowing the agent to initiate fibrosis inhibition.
In yet another aspect, anti-scarring agent may be located within pores or voids of the implantable sensor or implantable pump. For example, a implantable sensors and implantable pumps may be constructed to have cavities (e.g., divets or holes), grooves, lumen(s), pores, channels, and the like, which form voids or pores in the body of the implantable sensor or implantable pump. These voids may be filled (partially or completely) with a fibrosis- inhibiting agent or a composition that comprises a fibrosis-inhibiting agent.
In another aspect, an implantable sensor or implantable pump may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug. The reservoirs may be formed from divets in the device surface or micropores or channels in the device body. In one aspect, the reservoirs are formed from voids in the structure of the device. The reservoirs may house a single type of drug or more than one type of drug. The drug(s) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs. The filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier. In certain embodiments, the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount (dose) of drug, and each layer may have a different composition to further tailor the amount of drug that is released from the substrate. The multi-layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void. Within certain embodiments of the invention, the therapeutic compositions may also comprise additional ingredients such as surfactants (e.g., PLURONICS, such as F-127, L-122, L-101 , L-92, L-81 , and L-61), antiinflammatory agents (e.g., dexamethasone or asprin), anti-thrombotic agents (e.g., heparin, high activity heparin, heparin quaternary amine complexes (e.g., heparin benzalkonium chloride complex)), anti-infective agents (e.g., 5- fluorouracil, triclosan, rifamycim, and silver compounds), preservatives, antioxidants and/ or anti-platelet agents.
Within certain embodiments of the invention, the device or therapeutic composition can also comprise radio-opaque, echogenic materials and magnetic resonance imaging (MRI) responsive materials (i.e., MRI contrast agents) to aid in visualization of the device under ultrasound, fluoroscopy and/or MRI. For example, a device may be made with or coated with a composition which is echogenic or radiopaque (e.g., made with echogenic or radiopaque with materials such as powdered tantalum, tungsten, barium carbonate, bismuth oxide, barium sulfate, metrazimide, iopamidol, iohexol, iopromide, iobitridol , iomeprol , iopentol, ioversol, ioxilan, iodixanol, iotrolan, acetrizoic acid derivatives, diatrizoic acid derivatives, iothalamic acid derivatives , ioxithalamic acid derivatives, metrizoic acid derivatives, iodamide, lypophylic agents, iodipamide and ioglycamic acid or, by the addition of microspheres or bubbles which present an acoustic interface). Visualization of a device by ultrasonic imaging may be achieved using an echogenic coating. Echogenic coatings are described in, e.g., U.S. Patent Nos. 6,106,473 and 6,610,016. For visualization under MRI, contrast agents (e.g., gadolinium (III) chelates or iron oxide compounds) may be incorporated into or onto the device, such as, for example, as a component in a coating or within the void volume of the device (e.g., within a lumen, reservoir, or within the structural material used to form the device). In some embodiments, a medical device may include radio-opaque or MRI visible markers (e.g., bands) that may be used to orient and guide the device during the implantation procedure.
In another embodiment, these agents can be contained within the same coating layer as the therapeutic agent or they may be contained in a coating layer (as described above) that is either applied before or after the therapeutic agent containing layer.
Implantable pumps and sensor may, alternatively, or in addition, be visualized under visible light, using fluorescence, or by other spectroscopic means. Visualization agents that can be included for this purpose include dyes, pigments, and other colored agents. In one aspect, the medical implant may further include a colorant to improve visualization of the implant in vivo and/or ex vivo. Frequently, implants can be difficult to visualize upon insertion, especially at the margins of implant. A coloring agent can be incorporated into a medical implant to reduce or eliminate the incidence or severity of this problem. The coloring agent provides a unique color, increased contrast, or unique fluorescence characteristics to the device. In one aspect, a solid implant is provided that includes a colorant such that it is readily visible (under visible light or using a fluorescence technique) and easily differentiated from its implant site. In another aspect, a colorant can be included in a liquid or semisolid composition. For example, a single component of a two component mixture may be colored, such that when combined ex-vivo or in-vivo, the mixture is sufficiently colored.
The coloring agent may be, for example, an endogenous compound (e.g., an amino acid or vitamin) or a nutrient or food material and may be a hydrophobic or a hydrophilic compound. Preferably, the colorant has a very low or no toxicity at the concentration used. Also preferred are colorants that are safe and normally enter the body through absorption such as β- carotene. Representative examples of colored nutrients (under visible light) include fat soluble vitamins such as Vitamin A (yellow); water soluble vitamins such as Vitamin B12 (pink-red) and folic acid (yellow-orange); carotenoids such as β-carotene (yellow-purple) and lycopene (red). Other examples of coloring agents include natural product (berry and fruit) extracts such as anthrocyanin (purple) and saffron extract (dark red). The coloring agent may be a fluorescent or phosphorescent compound such as α-tocopherolquinol (a Vitamin E derivative) or L-tryptophan. Derivatives, analogues, and isomers of any of the above colored compound also may be used. The method for incorporating a colorant into an implant or therapeutic composition may be varied depending on the properties of and the desired location for the colorant. For example, a hydrophobic colorant may be selected for hydrophobic matrices. The colorant may be incorporated into a carrier matrix, such as micelles. Further, the pH of the environment may be controlled to further control the color and intensity.
In one aspect, the devices and composition of the present invention may include one or more coloring agents, also referred to as dyestuffs, which will be present in an effective amount to impart observable coloration to the composition, e.g., the gel. Examples of coloring agents include dyes suitable for food such as those known as F. D. & C. dyes and natural coloring agents such as grape skin extract, beet red powder, beta carotene, annato, carmine, turmeric, paprika, and so forth. Derivatives, analogues, and isomers of any of the above colored compound also may be used. The method for incorporating a colorant into an implant or therapeutic composition may be varied depending on the properties of and the desired location for the colorant. For example, a hydrophobic colorant may be selected for hydrophobic matrices. The colorant may be incorporated into a carrier matrix, such as micelles. Further, the pH of the environment may be controlled to further control the color and intensity.
In one aspect, the devices and compositions of the present invention include one or more preservatives or bacteriostatic agents, present in an effective amount to preserve the composition and/or inhibit bacterial growth in the composition, for example, bismuth tribromophenate, methyl hydroxybenzoate, bacitracin, ethyl hydroxybenzoate, propyl hydroxybenzoate, erythromycin, 5-fluorouracil, methotrexate, doxorubicin, mitoxantrone, rifamycin, chlorocresol, benzalkonium chlorides, and the like. Examples of the preservative include paraoxybenzoic acid esters, chlorobutanol, benzylalcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid, etc. In one aspect, the compositions of the present invention include one or more bactericidal (also known as bacteriacidal) agents. In one aspect, the devices and compositions of the present invention include one or more antioxidants, present in an effective amount. Examples of the antioxidant include sulfites, alpha-tocopherol and ascorbic acid. Within certain aspects of the present invention, the devices and therapeutic compositions of the present invention should be biocompatible, and release one or more fibrosis-inhibiting agents over a period of several hours, days, or, months. As described above, "release of an agent" refers to any statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the compositions and/or remains active on the surface of (or within) the composition. The compositions of the present invention may release the anti-scarring agent at one or more phases, the one or more phases having similar or different performance (e.g., release) profiles. The therapeutic agent may be made available to the tissue at amounts which may be sustainable, intermittent, or continuous; in one or more phases; and/or rates of delivery; effective to reduce or inhibit any one or more components of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue).
Thus, release rate may be programmed to impact fibrosis (or scarring) by releasing anti-scarring agent at a time such that at least one of the components of fibrosis is inhibited or reduced. Moreover, the predetermined release rate may reduce agent loading and/or concentration as well as potentially providing minimal drug washout and thus, increases efficiency of drug effect. Any one of the at least one anti-scarring agents may perform one or more functions, including inhibiting the formation of new blood vessels (angiogenesis), inhibiting the migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), inhibiting the deposition of extracellular matrix (ECM), and inhibiting remodeling (maturation and organization of the fibrous tissue). In one embodiment, the rate of release may provide a sustainable level of the anti-scarring agent to the susceptible tissue site. In another embodiment, the rate of release is substantially constant. The rate may decrease and/or increase over time, and it may optionally include a substantially non-release period. The release rate may comprise a plurality of rates. In an embodiment, the plurality of release rates may include rates selected from the group consisting of substantially constant, decreasing, increasing, and substantially non-releasing.
The total amount of anti-scarring agent made available on, in or near the device may be in an amount ranging from about 0.01 μg (micrograms) to about 2500 mg (milligrams). Generally, the anti-scarring agent may be in the amount ranging from 0.01 μg to about 10 μg; or from 10 μg to about 1 mg; or from 1 mg to about 10 mg; or from 10 mg to about 100 mg; or from 100 mg to about 500 mg; or from 500 mg to about 2500 mg.
The surface area amount of anti-scarring agent on, in or near the device may be in an amount ranging from less than 0.01 μg to about 2500 μg per mm2 of device surface area. Generally, the anti-scarring agent may be in the amount ranging from less than 0.01 μg; or from 0.01 μg to about 10 μg; or from 10 μg to about 250 μg; or from 250 μg to about 2500 μg per mm2.
The anti-scarring agent that is on, in or near the device may be released from the composition in a time period that may be measured from the time of implantation, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 7 days; from 7 days to about 14 days; from 14 days to about 28 days; from 28 days to about 56 days; from 56 days to about 90 days; from 90 days to about 180 days.
The amount of anti-scarring agent released from the composition as a function of time may be determined based on the in vitro release characteristics of the agent from the composition. The in vitro release rate may be determined by placing the anti-scarring agent within the composition or device in an appropriate buffer such as 0.1 M phosphate buffer (pH 7.4)) at 37°C. Samples of the buffer solution are then periodically removed for analysis by HPLC, and the buffer is replaced to avoid any saturation effects.
Based on the in vitro release rates, the release of anti-scarring agent per day may range from an amount ranging from about 0.01 μg (micrograms) to about 2500 mg (milligrams). Generally, the anti-scarring agent that may be released in a day may be in the amount ranging from 0.01 μg to about 10 μg; or from 10 μg to about 1 mg; or from 1 mg to about 10 mg; or from 10 mg to about 100 mg; or from 100 mg to about 500 mg; or from 500 mg to about 2500 mg. In one embodiment, the anti-scarring agent is made available to the susceptible tissue site in a programmed, sustained, and/or controlled manner which results in increased efficiency and/or efficacy. Further, the release rates may vary during either or both of the initial and subsequent release phases. There may also be additional phase(s) for release of the same substance(s) and/or different substance(s).
Further, therapeutic compositions and devices of the present invention should preferably have a stable shelf-life of at least several months and be capable of being produced and maintained under sterile conditions. Many pharmaceuticals are manufactured to be sterile and this criterion is defined by the USP XXII <1211>. The term "USP" refers to U.S. Pharmacopeia (see www.usp.org, Rockville, MD). Sterilization may be accomplished by a number of means accepted in the industry and listed in the USP XXII <1211>, including gas sterilization, ionizing radiation or, when appropriate, filtration. Sterilization may be maintained by what is termed asceptic processing, defined also in USP XXII <1211>. Acceptable gases used for gas sterilization include ethylene oxide. Acceptable radiation types used for ionizing radiation methods include gamma, for instance from a cobalt 60 source and electron beam. A typical dose of gamma radiation is 2.5 MRad. Filtration may be accomplished using a filter with suitable pore size, for example 0.22 μm and of a suitable material, for instance polytetrafluoroethylene (e.g., TEFLON from E.I. DuPont De Nemours and Company, Wilmington, DE). In another aspect, the compositions and devices of the present invention are contained in a container that allows them to be used for their intended purpose, i.e., as a pharmaceutical composition. Properties of the container that are important are a volume of empty space to allow for the addition of a constitution medium, such as water or other aqueous medium, e.g., saline, acceptable light transmission characteristics in order to prevent light energy from damaging the composition in the container (refer to USP XXII <661>), an acceptable limit of extractables within the container material (refer to USP XXII), an acceptable barrier capacity for moisture (refer to USP XXII <671>) or oxygen. In the case of oxygen penetration, this may be controlled by including in the container, a positive pressure of an inert gas, such as high purity nitrogen, or a noble gas, such as argon.
Typical materials used to make containers for pharmaceuticals include USP Type I through III and Type NP glass (refer to USP XXII <661>), polyethylene, TEFLON, silicone, and gray-butyl rubber.
In one embodiment, the product containers can be thermoformed plastics. In another embodiment, a seconday package can be used for the product. In another embodiment, product can be in a sterile container that is placed in a box that is labeled to describe the contents of the box.
Coating Implantable Sensors and Pumps with Fibrosis-lnhibiting
Agents
As described above, a range of polymeric and non-polymeric materials can be used to incorporate the fibrosis-inhibiting agent onto or into an implantable sensor or implantable pump. Coating the implantable sensor or implantable pump with these fibrosis-inhibiting agent-containing compositions, or with the fibrosis-inhibiting agent only, is one process that can be used to incorporate the fibrosis-inhibiting agent into or onto the implantable sensor or implantable pump. a. Dip coating
Dip coating is an example of coating process that can be used to associate the anti-scarring agent with the implantable sensor or implantable pump. In one embodiment, the fibrosis-inhibiting agent is dissolved in a solvent for the fibrosis-inhibiting agent and is then coated onto the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port).
Fibrosis-inhibiting Agent with an Inert Solvent In one embodiment, the solvent is an inert solvent for the implantable sensor or implantable pump such that the solvent does not dissolve the implantable device to any great extent and is not absorbed by the implantable device to any great extent. The implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time. The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The implantable sensor or implantable pump can then be removed from the solution. The rate at which the implantable sensor or implantable pump is withdrawn from the solution can be altered [e.g., 0.001 cm per sec to 50 cm per sec). The coated implantable sensor or implantable pump can be air-dried. The dipping process can be repeated one or more times depending on the specific application, where higher repetitions generally increase the amount of agent that is coated onto the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being coated on the surface of the device. Fibrosis-lnhibitinq Agent with a Swelling Solvent
In one embodiment, the solvent is one that will not dissolve the implantable sensor or implantable pump but will be absorbed by the device (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). In certain cases, these solvents can swell the implantable sensor or implantable pump to some extent. The implantable sensor or implantable pump can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time (seconds to days). The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The implantable sensor or implantable pump can then be removed from the solution. The rate at which the implantable sensor or implantable pump is withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated implantable sensor or implantable pump can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The fibrosis-inhibiting agent may also be present on the surface of the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated implantable sensor or implantable pump into a solvent for the fibrosis- inhibiting agent, or by spraying the implantable sensor or implantable pump with a solvent for the fibrosis-inhibiting agent. Fibrosis-lnhibiting Agent with a Solvent
In one embodiment, the solvent is one that will be absorbed by the implantable sensor or implantable pump and that will dissolve the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The implantable sensor or implantable pump can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time (seconds to hours). The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The implantable sensor or implantable pump can then be removed from the solution. The rate at which the implantable sensor or implantable pump is withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated implantable sensor or implantable pump can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) as well as being surface associated. Preferably, the exposure time of implantable sensor or implantable pump to the solvent does not incur significant permanent dimensional changes to the device (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The fibrosis-inhibiting agent may also be present on the surface of the implantable sensor and implantable pump. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the implantable sensor or implantable pump into a solvent for the fibrosis-inhibiting agent or by spraying the coated implantable sensor or implantable pump with a solvent for the fibrosis-inhibiting agent. In one embodiment, the fibrosis-inhibiting agent and a polymer are dissolved in a solvent, for both the polymer and the fibrosis-inhibiting agent, and are then coated onto the implantable sensor and implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port).
In the above description the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be one that has not been modified or one that has been further modified by coating with a polymer, surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In any one the above dip coating methods, the surface of the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be treated with a plasma polymerization method prior to coating of the fibrosis-inhibiting agent or fibrosis-inhibiting agent-containing composition, such that a thin polymeric layer is deposited onto the implantable sensor or implantable pump surface. Examples of such methods include parylene coating of devices and the use of various monomers such hydrocyclosiloxane monomers. Parylene coating may be especially advantageous if the device, or portions of the device (such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port), are composed of materials (e.g., stainless steel, nitinol) that do not allow incorporation of the therapeutic agent(s) into the surface layer using one of the above methods. A parylene primer layer may be deposited onto the implantable sensor or implantable pump using a parylene coater (e.g., PDS 2010 LABCOTER2 from Cookson Electronics) and a suitable reagent (e.g., di-. p-xylylene or dichloro-di-p-xylylene) as the coating feed material. Parylene compounds are commercially available, for example, from Specialty Coating Systems, Indianapolis, IN), including PARYLENE N (di-p-xylylene), PARYLENE C (a monchlorinated derivative of Parylene N, and PARYLENE D, a dichlorinated derivative of PARYLENE N).
b. Spray Coating Implantable Sensors and Implantable
Pumps Spray coating is another coating process that can be used. In the spray coating process, a solution or suspension of the fibrosis-inhibiting agent, with or without a polymeric or non-polymeric carrier, is nebulized and directed to the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) to be coated by a stream of gas. One can use spray devices such as an air-brush (for example models 2020, 360, 175, 100, 200, 150, 350, 250, 400, 3000, 4000, 5000, 6000 from Badger Air-brush Company, Franklin Park, IL), spray painting equipment, TLC reagent sprayers (for example Part # 14545 and 14654, Alltech Associates, Inc. Deerfield, IL, and ultrasonic spray devices (for example those available from Sono-Tek, Milton, NY). One can also use powder sprayers and electrostatic sprayers.
In one embodiment, the fibrosis-inhibiting agent is dissolved in a solvent for the fibrosis agent and is then sprayed onto the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port).
Fibrosis-lnhibitinq Agent with an Inert Solvent
In one embodiment, the solvent is an inert solvent for the implantable sensor or implantable pump such that the solvent does not dissolve the medical implantable sensor or implantable pump to any great extent and is not absorbed to any great extent. The implantable sensor or implantable pump can be held in place or mounted onto a mandrel or rod that has the ability to move in an X, Y or Z plane or a combination of these planes. Using one of the above described spray devices, the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be spray coated such that it is either partially or completely coated with the fibrosis- inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 ml_ per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated implantable sensor or implantable pump can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being coated on the surface of the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port).
Fibrosis-inhibiting Agent with a Swelling solvent
In one embodiment, the solvent is one that will not dissolve the implantable sensor or implantable pump but will be absorbed by it. These solvents can thus swell the implantable sensor or implantable pump to some extent. The implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated implantable sensor or implantable pump can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The fibrosis-inhibiting agent may also be present on the surface of the implantable sensor or implantable pump. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated implantable sensor or implantable pump into a solvent for the fibrosis-inhibiting agent, or by spraying the coated implantable sensor or implantable pump with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting Agent with a Solvent In one embodiment, the solvent is one that will be absorbed by the implantable sensor or implantable pump and that will dissolve it. The implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 ml_ per sec to 10 ml_ per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated implantable sensor or implantable pump can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) as well as being surface associated. In the preferred embodiment, the exposure time of the implantable sensor or implantable pump to the solvent may not incur significant permanent dimensional changes to it. The fibrosis-inhibiting agent may also be present on the surface of the implantable sensor or implantable pump. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated implantable sensor or implantable pump into a solvent for the fibrosis- inhibiting agent, or by spraying the coated implantable sensor or implantable pump with a solvent for the fibrosis-inhibiting agent.
In the above description the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be one that has not been modified as well as one that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In one embodiment, the fibrosis-inhibiting agent and a polymer are dissolved in a solvent, for both the polymer and the anti-fibrosing agent, and are then spray coated onto the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port).
Fibrosis-inhibiting Aqent/Polvmer with an Inert Solvent
In one embodiment, the solvent is an inert solvent for the implantable sensor or implantable pump such that the solvent does not dissolve it to any great extent and is not absorbed by it to any great extent. The implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/polymer/solvent solution for a specific period of time. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 ml_ per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated implantable sensor or implantable pump can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated on the surface of the device (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port).
Fibrosis-lnhibitino Agent/Polymer with a Swelling Solvent
In one embodiment, the solvent is one that will not dissolve the implantable sensor or implantable pump but will be absorbed by it. These solvents can thus swell the implantable sensor or implantable pump to some extent. The implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/polymer/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 ml_ per sec to 10 ml_ per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated implantable sensor or implantable pump can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated onto the surface of the implantable sensor or implantable pump as well as the potential for the fibrosis-inhibiting agent being adsorbed into the medical device (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated implantable sensor or implantable pump into a solvent for the fibrosis-inhibiting agent or by spraying the coated implantable sensor or implantable pump with a solvent for the fibrosis-inhibiting agent. Fibrosis-lnhibiting Agent/Polymer with a Solvent
In one embodiment, the solvent is one that will be absorbed by the implantable sensor or implantable pump and that will dissolve it. The implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated implantable sensor or implantable pump can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The implantable sensor or implantable pump can be dried under vacuum to reduce residual solvent levels. In the preferred embodiment, the exposure time of the implantable sensor or implantable pump to the solvent may not incur significant permanent dimensional changes to it (other than those associated with the coating itself). The fibrosis-inhibiting agent may also be present on the surface of the device (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port). The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated implantable sensor or implantable pump into a solvent for the fibrosis-inhibiting agent or by spraying the coated implantable sensor or implantable pump with a solvent for the fibrosis-inhibiting agent. In the above description the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) can be one that has not been modified as well as one that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process. In another embodiment, a suspension of the fibrosis-inhibiting agent in a polymer solution can be prepared. The suspension can be prepared by choosing a solvent that can dissolve the polymer but not the fibrosis- inhibiting agent, or a solvent that can dissolve the polymer and in which the fibrosis-inhibiting agent is above its solubility limit. In similar processes described above, the suspension of the fibrosis-inhibiting and polymer solution can be sprayed onto the implantable sensor or implantable pump (or part of the sensor or pump such as the body, the detector, the semipermeable membrane, the drug delivery catheter, or the drug delivery port) such that it is coated with a polymer that has a fibrosis-inhibiting agent suspended within it.
2. Systemic, Regional and Local Delivery of Fibrosis-inhibiting Agents
A variety of drug-delivery technologies are available for systemic, regional and local delivery of fibrosis-inhibiting therapeutic agents. Several of these techniques may be suitable to achieve preferentially elevated levels of fibrosis-inhibiting agents in the vicinity of the implantable sensors and implantable pump, including: (a) using drug-delivery catheters for local, regional or systemic delivery of fibrosis-inhibiting agents to the tissue surrounding the device or implant. Typically, drug delivery catheters are advanced through the circulation or inserted directly into tissues under radiological guidance until they reach the desired anatomical location. The fibrosis-inhibiting agent can then be released from the catheter lumen in high local concentrations in order to deliver therapeutic doses of the drug to the tissue surrounding the device or implant; (b) drug localization techniques such as magnetic, ultrasonic or MRI-guided drug delivery; (c) chemical modification of the fibrosis-inhibiting drug or formulation designed to increase uptake of the agent into damaged tissues (e.g., antibodies directed against damaged or healing tissue components such as macrophages, neutrophils, smooth muscle cells, fibroblasts, extracellular matrix components, neovascular tissue); (d) chemical modification of the fibrosis-inhibiting drug or formulation designed to localize the drug to areas of bleeding or disrupted vasculature; and/or (e) direct injection or administration of the fibrosis-inhibiting agent, for example, under endoscopic vision.
3. Infiltration of Fibrosis-inhibiting Agents into the Tissue
Surrounding a Device or Implant Alternatively, the tissue surrounding the implantable sensor or implantable pump can be treated with a fibrosis-inhibiting agent or a composition prior to, during, or after the implantation procedure. A fibrosis- inhibiting agent or a composition comprising a fibrosis-inhibiting agent may be infiltrated around the device or implant, for example, by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the medical device; (b) the vicinity of the medical device-tissue interface; (c) the region around the medical device; and (d) tissue surrounding the medical device.
Methods for infiltrating the subject polymer compositions into tissue adjacent to a medical device include delivering the fibrosis-inhibiting agent or composition: (a) to the medical device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the medical device; (c) to the surface of the medical device and/or the tissue surrounding the implanted medical device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the medical device; (d) by topical application of the composition into the anatomical space where the medical device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the medical device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (e.g., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject polymer compositions may be infiltrated into tissue adjacent to all or a portion of the device.
It may be noted that certain polymeric carriers themselves can help prevent the formation of fibrous tissue around the implantable sensors and implantable pumps. The following exemplary polymer compositions may be used for the practice of this embodiment, either alone, or in combination with a fibrosis inhibiting composition. The following polymeric carriers can be infiltrated (as described in the previous paragraph) into the vicinity of the device-tissue interface and include: (a) sprayable collagen-containing formulations such as COSTASIS and CT3, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (b) sprayable PEG-containing formulations such as COSEAL, FOCALSEAL , SPRAYGEL or DURASEAL, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (d) hyaluronic acid- containing formulations such as RESTYLANE, HYLAFORM, PERLANE,
SYNVISC, SEPRAFILM, SEPRACOAT, loaded with a fibrosis-inhibiting agent applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (e) polymeric gels for surgical implantation such as REPEL or FLOWGEL loaded with a fibrosis-inhibiting agent applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (f) orthopedic "cements" used to hold prostheses and tissues in place loaded with a fibrosis-inhibiting agent applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface), such as OSTEOBOND, low viscosity cement (LVC), SIMPLEX P, PALACOS, and ENDURANCE; (g) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE® SOOTHE-N-SEAL LIQUID PROTECTANT, either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (h) implants containing hydroxyapatite (or synthetic bone material such as calcium sulfate, VITOSS and CORTOSS) loaded with a fibrosis-inhibiting agent applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (i) other biocompatible tissue fillers loaded with a fibrosis-inhibiting agent, such as those made by BioCure, Inc., 3M Company and Neomend, Inc., applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); (j) polysaccharide gels such as the ADCON series of gels either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface); and/or (k) films, sponges or meshes such as INTERCEED, VICRYL mesh, and GELFOAM loaded with a fibrosis-inhibiting agent applied to the implantation site (or the device, detector, semipermeable membrane, drug delivery catheter, and/or drug delivery port surface).
An exemplary polymeric matrix useful in preventing the formation of fibrous tissue around the implantable sensor or implantable pump, either alone or in combination with a fibrosis (or gliosis) inhibiting agent/composition, may be formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another exemplary composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue around the implantable sensor or implantable pump.
Other examples of polymer compositions that may be infiltrated into tissue adjacent to a medical device include compositions formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another exemplary composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix.
Implantable Sensors
In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to an implantable sensor. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of implantable sensors that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with implantable sensors have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Implantable sensors are provided that can be used to detect physiological levels or changes in the body. There are numerous sensor devices where the occurrence of a fibrotic reaction will adversely affect the functioning of the device or the biological problem for which the device was implanted or used. Proper clinical functioning of an implanted sensor is dependent upon intimate anatomical contact with the target tissues and/or body fluids. Scarring around the implanted device may degrade the electrical components and characteristics of the device-tissue interface, and the device may fail to function properly. The formation of scar tissue between the sensing device and the adjacent (target) tissue can prevent the flow of physical, chemical and/or biological information (e.g., fluid levels, drug levels, metabolite levels, glucose levels, pressure etc.) from reaching the detection mechanism of the sensor. Similarly if a "foreign body" response occurs and causes the implanted sensor to become encapsulated by scar (i.e., the body "walls off' the sensor with fibrous tissue), the sensor will receive biological information that is not reflective of the organism as a whole. If the sensor is detecting conditions inside the capsule (i.e., levels detected in a microenvironment), and these conditions are not consistent with those outside the capsule (i.e., within the body as a whole - the microenvironment), it will record information that is not representative of systemic levels. Implantation of an implantable sensor may also introduce or promote infection in the vicinity of the implant site.
Sensors or transducers may be located deep within the body for monitoring a variety of physiological properties, such as temperature, pressure, strain, fluid flow, metabolite levels (e.g., electrolytes, glucose), drug levels, chemical properties, electrical properties, magnetic properties, and the like. Representative examples of implantable sensors for use in the practice of the invention include, blood and tissue glucose monitors, electrolyte sensors, blood constituent sensors, temperature sensors, pH sensors, optical sensors, amperometric sensors, pressure sensors, biosensors, sensing transponders, strain sensors, activity sensors and magnetoresistive sensors.
Numerous types of implantable sensors and transducers have been described. For example, the implantable sensor may be a micro- electronic device that is implanted around the large bowels to control bowel function by detecting rectal contents and stimulating peristaltic contractions to empty the bowels when it is convenient. See, e.g., U.S. Patent No. 6,658,297. The implantable sensor may be used to measure pH in the Gl tract. A representative example of such a pH sensing device is the BRAVO pH Monitoring System from Medtronic, Inc. (Minneapolis, MN). The implantable sensor may be part of a Gl catheter or probe that includes a sensor portion connected to an electrical or optical measurement device and a sensitive polymeric material that undergoes an irreversible change when exposed to cumulative action of an external medium. See, e.g., U.S. Patent No. 6,006,121. The implantable sensor may be a component of a central venous catheter (CVC) (e.g., a jugular vein catheter) system. For example, the device may be composed of a catheter body having at least one oxygen sensor and a distal heat exchange region in which the catheter body is formed with coolant supply and return lumens to provide heat exchange within a body to prevent overheating due to severe brain trauma or ischemia due to stroke. See, e.g., U.S. Patent No. 6,652,565. A CVC may include a thermal mass and a temperature sensor to measure blood temperature. See, e.g., U.S. Patent No. 6,383,144.
In one aspect, the present invention provides implantable sensors having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with implantable sensors have been described above.
Agents or compositions of the present invention may be infiltrated around implanted implantable sensors by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the implantable sensor; (b) the vicinity of the implantable sensor-tissue interface; (c) the region around the implantable sensor; and (d) tissue surrounding the implantable sensor. Methods for infiltrating the subject compositions into tissue adjacent to an implantable sensor include delivering the agent or composition: (a) to the surface of the implantable sensor (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the implantable sensor; (c) to the surface of the implantable sensor and/or the tissue surrounding the implanted implantable sensor (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the implantable sensor; (d) by topical application of the composition into the anatomical space where the implantable sensor may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the implantable sensor as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, sensor only, detector only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to implantable sensors may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin; radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, juglone, tacrolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As implantable sensors are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2 Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 -40 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC5O range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1 % of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10~6 about 10"6 to 10'5 or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Several specific implantable sensor devices and treatments will be described in greater detail below.
(1) Blood and Glucose Monitors In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to a glucose monitor. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti- scarring and/or anti-infective agent).
Glucose monitors are used to detect changes in blood glucose, specifically for the management and treatment of patients with diabetes mellitus. Diabetes is a metabolic disorder of glucose metabolism that afflicts tens of millions of people in the developed countries of the world. This disease is characterized by the inability of the body to properly utilize and metabolize carbohydrates, particularly glucose. Normally, the finely-tuned balance between glucose in the blood and glucose in the bodily tissue cells is maintained by insulin, a hormone produced by the pancreas. If the pancreas becomes defective and insulin is produced in inadequate amounts to reduce blood glucose levels (Type I diabetes), or if the body becomes insensitive to the glucose-lowering effects of insulin despite adequate pancreatic insulin production (Type Il diabetes), the result is diabetes. Accurate detection of blood glucose levels is essential to the management of diabetic patients because the dosage and timing of administration of insulin and/or other hypoglycemic agents are titrated depending upon changes in glucose levels in response to the medication. If the dosage is too high, blood glucose levels drop too low, resulting in confusion and potentially even loss of consciousness. If the dosage is too low, blood glucose levels rise too high, leading to excessive thirst, urination, and changes in metabolism known as ketoacidosis. If the timing of medication administration is incorrect, blood glucose levels can fluctuate wildly between the two extremes - a situation that is thought to contribute to some of the long-term complications of diabetes such as heart disease, kidney failure and blindness. Since in the extreme, all these conditions can be life threatening, careful and continuous monitoring of glucose levels is a critical aspect of diabetes management. One way to detect changes in glucose levels and to continuously sense when levels of glucose become too high or too low in diabetes patients is to implant a glucose monitor. As the glucose monitor detects changes in the blood glucose levels, insulin can be administered by external injection or via an implantable insulin pump to maintain blood glucose levels within an acceptable physiologic range.
Numerous types of blood and tissue glucose monitors are suitable for use in the practice of the invention. For example, the glucose monitor may be delivered to the vascular system transluminal^ using a catheter on a stent platform. See, e.g., U.S. Patent No. 6,442,413. The glucose monitor may be composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations. See, e.g., U.S. Patent Nos. 5,101 ,814 and 5,190,041. The glucose monitor may be a small diameter flexible electrode implanted subcutaneously which may be composed of an analyte-responsive enzyme designed to be an electrochemical glucose monitor. See, e.g., U.S. Patent Nos. 6,121 ,009 and 6,514,718. The implantable sensor may be a closed loop insulin delivery system whereby there is a sensing means that detects the patient's blood glucose level based on electrical signals and then stimulates either an insulin pump or the pancreas to supply insulin. See, e.g., U.S. Patent Nos. 6,558,345 and 6,093,167. Other glucose monitors are described in, for e.g., U.S. Patent Nos. 6,579,498; 6,565,509 and 5,165,407. Minimally invasive glucose monitors include the GLUCOWATCH G2 BIOGRAPHER from Cygnus Inc. (see cygn.com); see, e.g., U.S. Patent Nos. 6,546,269; 6,687,522; 6,595,919 and U.S. Patent Application Nos. 20040062759A1; 20030195403A1 ; and 20020091312A1.
Glucose monitors, which may benefit from having the subject composition infiltrated into adjacent tissue according to the present invention, include commercially available products. Numerous commercially available blood and tissue glucose monitoring devices are suitable for the practice of this invention. Although virtually any implantable glucose monitor may be utilized, several specific commercial and development stage examples are described below for greater clarity.
The CONTINUOUS GLUCOSE MONITORING SYSTEM (CGMS) from Medtronic MiniMed, Inc. (Northridge, CA; see minimed.com); see, e.g., U.S. Patent Nos. 6,520,326; 6,424,847; 6,360,888; 5,605,152; 6,804,544; and U.S. Patent Application No. 20040167464A1. The CGMS system is surgically implanted in the subcutaneous tissue of the abdomen and stores tissue glucose readings every 5 minutes. Infiltrating the subject composition into tissue adjacent to the sensor may prolong the activity of this device because it often must be removed after several days (approximately 3), in part because it loses its sensitivity as a result of the local tissue reaction to the device.
The CONTINUOUS GLUCOSE MONITORING DEVICE from TheraSense (Alameda, CA, see therasense.com) which utilizes a disposable, miniaturized electrochemical sensor that is inserted under the patient's skin using a spring-loaded insertion device. The sensor measures glucose levels in the interstitial fluid every five minutes, with the ability to store results for future analysis. See, e.g., US20040186365A1 ; US20040106858A1 and US20030176183A1. Even though the device can store up to a month of data and has alarms for high and low glucose levels, it must be replaced every few days because it loses its accuracy as a result of the foreign body reaction to the implant. Infiltrating the subject composition into tissue adjacent to this sensor may prolong its activity, enhance its performance and reduce the frequency of replacement. Another electrochemical sensor that may benefit from the present invention is the multilayered implantable electrochemical sensor from lsense (Portland, OR). This system consists of a semipermeable membrane, a catalytic membrane which generates an electrical current in the presence of glucose, and a specificity membrane to reduce interference from other substances.
The SMSI glucose sensor (Sensors for Medicine and Sciences, Inc., Montgomery County, Maryland; see s4ms.com) is designed to be implanted under the skin in a short outpatient procedure. The sensor is designed to automatically measure interstitial glucose every few minutes, without any user intervention. The sensor implant communicates wirelessly with a small external reader, allowing the user to monitor glucose levels continuously or on demand. The reader is designed to be able to track the rate of change of glucose levels and warn the user of impending hypo- or hyperglycemia. The operational life of the sensor implant is about 6-12 months, after which it may be replaced.
Animas Corporation (West Chester, PA; animascorp.com) is developing an implantable glucose sensor that measures the near-infrared absorption of blood based on spectroscopy or optical sensing placed around a vein. The Animas glucose monitor may be tied to an insulin infusion pump to provide a closed-loop control of blood glucose levels. Scar tissue over the sensor distorts the ability of the device to correctly gather optical information and the sensor may thus benefit from the present invention. DexCom, Inc. (San Diego, CA; see dexcom.com) is developing their Continuous Glucose Monitoring System which is an implantable sensor that wirelessly transmits continuous blood glucose readings to an external receiver. The receiver displays the current glucose value every 30 seconds, as well as one-hour, three-hour and nine-hours trended values, and sounds an alert when a high or low glucose excursion is detected. This device features an implantable sensor that is placed in the subcutaneous tissue and continuously monitors tissue (interstitial fluid) glucose levels for both type 1 and type 2 diabetics. This device may also include a unique microarchitectural arrangement in the sensor region that allows accurate data to be obtained over long periods of time. Glucose monitoring devices and associated systems that are developed by DexCom, Inc. are described in, for example, U.S. Patent Nos. 6,741,877; 6,702,857 and 6,558,321. Unfortunately, even though the battery and circuitry of monitoring devices allows long-term functioning, a foreign body response and/or encapsulation of the implant affect the ability of the device to detect glucose levels accurately for prolonged periods in a percentage of implants. Infiltrating the subject composition into tissue adjacent to this device may allow it to accurately detect glucose levels for longer periods of time after implantation, reduce the number of devices that fail and decrease the incidence of replacement.
Also of particular interest in the practice of this invention is glucose monitoring systems that utilize a glucose-responsive polymer as part of their detection mechanism. M-Biotech (Salt Lake City, UT) is developing a continuous monitoring system that consists of subcutaneous implantation of a glucose-responsive hydrogel combined with a pressure transducer. See, e.g., U.S. Patent Nos.; and. The hydrogel responds to changes in glucose concentration by either shrinking or swelling and the expansion or contraction is detected by the pressure transducer. The transducer converts the information into an electrical signal and sends a wireless signal to a display device. Cybersensors (Berkshire, UK) produces a capsule-like sensor implanted under the skin and an external receiver/transmitter that captures the data and powers the capsule via RF signals (see, e.g., GB 2335496 and U.S. Patent No. 6,579,498) Issued by the UK Patent and Trademark Office). The sensor capsule is composed of a glucose affinity polymer and contains a physical sensor and an RF microchip; the entire capsule is further enclosed in a semipermeable membrane. The glucose affinity polymer exhibits rheological changes when exposed to glucose (in the range of 3-15 nM) by becoming thinner and less viscous as glucose concentrations increase. This reversible reaction can be detected by the physical sensor and converted into a signal. These aforementioned systems are suitable for infiltrating the subject composition into tissue adjacent to the implanted sensor as provided in the present invention. Another glucose sensing device is under development by
Advanced Biosensors (Mentor, OH) that consists of small (150 μm wide by 2 mm long), biocompatible, silicon-based needles that are implanted under the skin. The device senses glucose levels in the dermis and transmits data wirelessly. Unfortunately, a foreign body response and/or encapsulation of the implant affect the ability of the device to detect glucose levels accurately for longer than 7 days. Infiltrating the subject composition into tissue adjacent to this device may allow it to accurately detect glucose levels for longer periods of time and extend the effective lifespan of the device.
Regardless of the specific design features of implantable blood, tissue, or interstitial fluid glucose monitoring devices, for accurate detection of physical, chemical and/or physiological properties, the device must be accurately positioned adjacent to the tissue. In particular, the detector of the sensing mechanism must be exposed to glucose levels that are identical to (or representative of) those found in the bloodstream. If excessive scar tissue growth or extracellular matrix deposition occurs around the device, this can impair the movement of glucose from the tissue to the detector and render it ineffective. Similarly if a "foreign body" response occurs and causes the implanted glucose sensor to become encapsulated by fibrous tissue, the sensor will be detecting glucose levels in the capsule. If glucose levels inside the capsule are not consistent with those outside the capsule (i.e., within the body as a whole), it will record information that is not representative of systemic levels. This can cause the physician or the patient to administer the wrong dosage of hypoglycemic drugs (such as insulin) with potentially serious consequences. Blood, tissue or interstitial fluid glucose monitoring devices having the subject compositions infiltrated into tissue adjacent to the implant can reduce scarring and/or encapsulation of the implant and increase the efficiency and accuracy of glucose detection, minimize insulin dosing errors, assist in the maintenance of correct blood glucose levels, increase the duration that these devices function clinically, and/or reduce the frequency of implant replacement. Glucose monitoring devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. In one aspect, the device includes blood, tissue and interstitial fluid glucose monitoring devices having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the device is or will be implanted. In another aspect, the present invention provides glucose monitoring devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with glucose monitoring devices have been described above. Agents or compositions of the present invention may be infiltrated around implanted glucose monitoring devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the glucose monitoring device; (b) the vicinity of the glucose monitoring device-tissue interface; (c) the region around the glucose monitoring device; and (d) tissue surrounding the glucose monitoring device. Methods for infiltrating the subject compositions into tissue adjacent to a glucose monitoring device include delivering the composition: (a) to the surface of the glucose monitoring device (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the glucose monitoring device; (c) to the surface of the glucose monitoring device and/or the tissue surrounding the implanted glucose monitoring device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the glucose monitoring device; (d) by topical application of the composition into the anatomical space where the glucose monitoring device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the glucose monitoring device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, sensor only, detector only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to glucose monitoring devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced. Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As glucose monitoring devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti- scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC5Q range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM ICs0 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10~8- 10"4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10'7 to 10"6 about 10~6 to 10"5 or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracϋ), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
(2) Pressure and Stress Sensors In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to a pressure and/or stress sensor. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Pressure or stress monitors may be used to detect increasing pressure or stress within the body. Implantable pressure transducers and sensors are used for temporary or chronic use in a body organ, tissue or vessel for recording absolute pressure. Many different designs and operating systems have been proposed and placed into temporary or chronic use for patients with a variety of medical conditions. Indwelling pressure sensors for temporary use of a few days or weeks are available, however, chronically or permanently implantable pressure sensors have also been used. Pressure sensors may detect many types of bodily pressures, such as, but not limited to blood pressure and fluid flow, pressure within aneurysm sacs, intracranial pressure, and mechanical pressure associated with bone fractures.
Numerous types of pressure monitors are suitable for use in the practice of the invention. For example, the implantable sensor may detect body fluid absolute pressure at a selected site and ambient operating temperature by using a lead, sensor module, sensor circuit (including electrical conductors) and means for providing voltage. See, e.g., U.S. Patent No. 5,535,752. The implantable sensor may be an intracranial pressure monitor that provides an analogue data signal which is converted electronically to a digital pulse. See, e.g., U.S. Patent No. 6,533,733. The implantable sensor may be a barometric pressure sensor enclosed in an air chamber which is used for deriving reference pressure data for use in combination with an implantable medical device, such as a pacemaker. See, e.g., U.S. Patent No. 6,152,885. The implantable sensor may be adapted to be inserted into a body passageway to monitor a parameter related to fluid flow through an endoluminal implant (e.g., stent). See, e.g., U.S. Patent No. 5,967,986. The implantable sensor may be a passive sensor with an inductor-capacitor circuit having a resonant frequency which is adapted for the skull of a patient to sense intracranial pressure. See, e.g., U.S. Patent No. 6,113,553. The implantable sensor may be a self- powered strain sensing system that generates a strain signal in response to stresses that may be produced at a bone fixation device. See, e.g., U.S. Patent No. 6,034,296. The implantable sensor may be a component of a perfusion catheter. The catheter may include a wire electrode and a lumen for perfusing saline around the wire, which is designed for measuring a potential difference across the Gl wall and for simultaneous measurement of pressure. See, e.g., U.S. Patent No. 5,551 ,425. The implantable sensor may be part of a CNS device; for example, an intracranial pressure sensor which is mounted within the skull of a body at the situs where the pressure is to be monitored and a means of transmitting the pressure externally from the skull. See, e.g., U.S. Patent No. 4,003,141. The implantable sensor may be a component of a left ventricular assist device. For example, the VAD may be a blood pump adapted to be joined in flow communication between the left ventricle and the aorta using an inlet flow pressure sensor and a controller that may adjust speed of pump based on sensor feedback. See, e.g., U.S. Patent No. 6,623,420.
Pressure and/or stress sensor devices, which may benefit from having the subject composition infiltrated into adjacent tissue according to the present invention, include commercially available products. Numerous commercially available and experimental pressure and stress sensor devices are suitable for the practice of the invention. By way of illustration, a selection of these devices and implants are described in the following paragraphs.
A device from CardioMEMS (Atlanta, GA; @cardiomems.com, a partnership between the Georgia Institute of Technology and the Cleveland Clinic) which can be inserted into an aneurysm sac to monitor pressure within the sac and thereby alert a medical specialist to the filing of the sac with fluid, possibly to rupture-provoking levels. Endovascular aneurysm repair (EVAR) is often performed using a stent graft which isolates the aneurysm from the circulation. However, persistent leakage of blood into the aneurysm sac results in ongoing pressure build-up in the sac and a resultant risk of rupture. The CardioMEMS device is implanted into the aneurysm sac after EVAR to monitor pressure in the isolated sac in order to detect which patients are at increasing risk of rupture. The pressure sensor features an inductive-capacitive resonant circuit with a variable capacitor. Since capacitance varies with the pressure in the environment in which the capacitor is placed, it can detect changes in local pressure. Data is generated by using external excitation systems that induce an oscillating current in the sensor and detecting the frequency of oscillation (which is then used to calculate pressure). Unfortunately, even though the circuitry allows long-term functioning, a foreign body response and/or encapsulation of the implant affect the ability of the device to detect accurate pressure levels in the aneurysm (i.e., the device detects the pressure in the microenvironment of the capsule, not of the aneurysm sac as a whole). Implantation of a sensor may also introduce or promote infection in the vicinity of the implant site. Infiltrating the subject composition into tissue adjacent to this device may allow it to accurately detect pressure levels for longer periods of time after implantation and reduce the number of devices that fail.
MicroStrain Inc. (Williston, VT, @microstrain.com) has developed a family of wireless implantable sensors for measuring strain, position and motion within the body. These sensors can measure, for example, eye tremor, depth of corneal implant, orientation sensor for improved tooth crown prep, mayer ligament strains, spinal ligament strains, vertebral bone strains, elbow ligament strains, emg and ekg data, 3DM-G for measurement of orientation and motion, wrist ligament strains, hip replacement sensors for measuring micromotion, implant subsidence, knee ligament strain, ankle ligament strain, Achilles tendon strain, foot arch support strains, force within foot insoles. The company provides a knee prosthesis that can measure in vivo compressive forces and transmit the data in real time. Patents describing this technology, and components used in the manufacture of devices for this technology include US 6,714,763; 6,625,517; 6,622,567; 6,588,282; 6,529,127; 6,499,368; 6,433,629; 5,887,351 ; 5,777,467; 5,497,147; and 4,993,428. US Patent Applications describing this technology, and components used in the manufacture of devices for this technology include 20040113790; 20040078662; 20030204361 ; 20030158699; 20030047002; 20020190785; 20020170193; 20020088110; 20020085174; 20010054317; and 20010033187. Mesotec (Hannover, Germany; @mesotec.com), in collaboration with several German institutes (e.g., Fraunhofer Institute of Microelectronic Circuits and Systems), has developed an implantable intraocular pressure sensor system, called the MESOGRAPH, which can continuously monitor intraocular pressure. This is desirable, e.g., in order to identify the onset of glaucoma. The CMOS-based sensor can be implanted during standard surgical procedures and is inductively linked to an external unit integrated into a spectacle frame. The glasses are in turn linked via a cable to a portable data logger. Data is relayed upstream to the glasses using a modulated RF carrier operating at 13.56 MHz and a switchable load, while power comes downstream to the sensor. By varying the diameter of the polysilicon diaphragms in the on- chip micromechanical vacuum gap capacitors, the pressure range to which the sensor responds can be adapted between 50kNm-2 and 3.5MNm-2. The device consists of a fine, foldable coil for telemetric coupling and a very small miniaturized pressure sensor. The sensor is manufactured on a micro- technological basis and serves for continuous, long-term reading and monitoring of intraocular pressure. Chip and coil are integrated in modified soft intraocular lenses, which can be implanted in the patient's eye during today's common surgical procedures. Unfortunately, the device often fails after initially successful implantation because a foreign body response and/or encapsulation of the implant affect the ability of it to detect accurate pressure levels in the eye (i.e., the device detects the pressure in the microenvironment of the capsule surrounding the implant, not intraocular pressure as a whole). Implantation of a sensor may also introduce or promote infection in the vicinity of the implant site. Infiltrating the subject composition into the eye tissue adjacent to this device may allow it to accurately detect pressure levels for longer periods of time after implantation and reduce the number of devices that fail.
Regardless of the specific design features of the pressure and/or stress sensor, for accurate detection of physical and/or physiological properties (such as pressure), the device must be accurately positioned within the tissue and receive information that is representative of conditions as a whole. If excessive scar tissue growth or extracellular matrix deposition occurs around the device, the sensor may receive erroneous information that compromises its efficacy or the scar tissue may block the flow of biological information to the sensor. For example, many devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant pressure levels (i.e., the device detects the pressure in the microenvironment of the capsule surrounding the implant, not the pressure of the larger environment). Pressure and stress sensing devices having the subject compositions infiltrated into tissue adjacent to the implant can increase the efficiency of detection and increase the duration that these devices function clinically. Pressure and stress sensing devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. In one aspect, the device includes implantable sensor devices having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the device is or will be implanted. In another aspect, the present invention provides pressure or stress sensing devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with pressure or stress sensing devices have been described above. Agents or compositions of the present invention may be infiltrated around implanted pressure or stress sensing devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the pressure or stress sensing device; (b) the vicinity of the pressure or stress sensing device-tissue interface; (c) the region around the pressure or stress sensing device; and (d) tissue surrounding the pressure or stress sensing device. Methods for infiltrating the subject compositions into tissue adjacent to a pressure or stress sensing device include delivering the composition: (a) to the surface of the pressure or stress sensing device (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the pressure or stress sensing device; (c) to the surface of the pressure or stress sensing device and/or the tissue surrounding the implanted pressure or stress sensing device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the pressure or stress sensing device; (d) by topical application of the composition into the anatomical space where the pressure or stress sensing device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the pressure or stress sensing device as a solution as an infusate or as a sustained release preparation; (T) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, sensor only, detector only and/or a combination thereof. According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to pressure or stress sensing devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As pressure or stress sensing devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti- scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2. Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10~8- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10'8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10"5or about 10"5 to 10"4of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
(3) Cardiac Sensors
In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to a cardiac sensor device. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). In another aspect, the implantable sensor may be a device configured to detect properties in the heart or in cardiac muscle tissue. Cardiac sensors are used to detect parameters associated with the performance of the heart as monitored at any given time point along a prolonged time period. Typically, monitoring of the heart is often conducted to detect changes associated with heart disease, such as chronic heart failure (CHF). By monitoring patterns associated with heart function, deterioration based on hemodynamic changes can be detected (parameters such as cardiac output, ejection fraction, pressure, ventricular wall motion, etc.). This constant direct monitoring is central to disease management in patients that present with CHF. By monitoring hemodynamic measures directly using implantable sensors, a hemodynamic crisis can be detected and the appropriate medications and interventions selected.
Numerous types of cardiac sensors are suitable for use in the practice of the invention. For example, the implantable sensor may be an activity sensor incorporating a magnet and a magnetoresistive sensor that provides a variable activity signal as part of a cardiac device. See, e.g., U.S. Patent No. 6,430,440 and 6,411,849. The implantable sensor may monitor blood pressure in a heart chamber by emitting wireless communication to a remote device. See, e.g., U.S. Patent No. 6,409,674. The implantable sensor may be an accelerometer-based cardiac wall motion sensor which transduces accelerations of cardiac tissue to a cardiac stimulation device by using electrical signals. See, e.g., U.S. Patent No. 5,628,777. The implantable sensor may be implanted in the heart's cavity with an additional sensor implanted in a blood vessel to detect pressure and flow within heart's cavity. See, e.g., U.S. Patent No. 6,277,078.
Cardiac sensors, which may benefit from having the subject composition infiltrated into adjacent tissue according to the present invention, include commercially available products. Commercially available cardiac sensor devices suitable for the practice of the invention include Biotronik's (Biotronik GmbH & Co., Berlin, Germany, see biotronik.com) CARDIAC
AIRBAG ICD SYSTEM is a rhythm monitoring device that offers rescue shock capability delivering 30 Joule shock therapies for up to 3 episodes of ventricular fibrillation. In addition to the rescue shock capability the system can also provide bradycardia pacing and VT monitoring. The PROTOS family of pacemakers from Biotronik (see biotronikusa.com) also incorporates pacing sensor capability called Closed Loop Simulation.
Blood flow and tissue perfusion monitors can be used to monitor noncardiac tissue as well. Researchers at Oak Ridge National Laboratory have developed a wireless sensor that monitors blood flow to a transplanted organ for the early detection of transplant rejection.
Medtronic (Minneapolis, MN; see medtronic.com) is developing their CHRONICLE implantable product, which is designed to continuously monitor a patient's intracardiac pressures, heart rate and physical activity using a sensor placed directly in the heart's chamber. The patient periodically downloads this information to a home-based device that transmits this physiologic data securely over the Internet to a physician.
Regardless of the specific design features of the cardiac sensor, for accurate detection of physical and/or physiological properties (such as pressure, flow rates, etc.), the device must be accurately positioned within the heart muscle, chambers or great vessels and receive information that is representative of conditions as a whole. If excessive scar tissue growth or extracellular matrix deposition occurs around the sensing device, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of biological information to the detector mechanism of the sensor. For example, many cardiac sensors fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (i.e., the device detects conditions in the microenvironment of the capsule surrounding the implant, not the pressure of the larger environment). Cardiac sensor devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. Cardiac sensing devices having the subject compositions infiltrated into tissue adjacent to the implant can increase the efficiency of detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the device is or will be implanted. In another aspect, the present invention provides cardiac sensing devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with cardiac sensing devices have been described above. Agents or compositions of the present invention may be infiltrated around implanted cardiac sensor devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the cardiac sensor device; (b) the vicinity of the cardiac sensor device-tissue interface; (c) the region around the cardiac sensor device; and (d) tissue surrounding the cardiac sensor device. Methods for infiltrating the subject compositions into tissue adjacent to a cardiac sensor device include delivering the composition: (a) to the surface of the cardiac sensor device (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the cardiac sensor device; (c) to the surface of the cardiac sensor device and/or the tissue surrounding the implanted cardiac sensor device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the cardiac sensor device; (d) by topical application of the composition into the anatomical space where the cardiac sensor device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the cardiac sensor device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, sensor only, detector only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to cardiac sensor devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced. Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As cardiac sensor devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10~8- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10'7 to 10"6 about 10'6 to 10"5 or about 10"5 to 10'4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
(4) Respiratory Sensors
In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to a respiratory sensor device. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
The implantable sensor may be a device configured to detect properties in the respiratory system. Respiratory sensors may be used to detect changes in breathing patterns. For example, a respiratory sensor may be used to detect sleep apnea, which is an airway disorder. There are two kinds of sleep apnea. In one condition, the body fails to automatically generate the neuromuscular stimulation necessary to initiate and control a respiratory cycle at the proper time. In the other condition, the muscles of the upper airway contract during the time of inspiration and thus the airway becomes obstructed. The cardiovascular consequences of apnea include disorders of cardiac rhythm (bradycardia, auriculoventricular block, ventricular extrasystoles) and hemodynamic disorders (pulmonary and systemic hypertension). This results in a stimulatory metabolic and mechanical effect on the autonomic nervous system and the potential to ultimately lead to increased morbidity. To treat this condition, implantable sensors may be used to monitor respiratory functioning to detect an apnea episode so the appropriate response (e.g., electrical stimulation to the nerves of the upper airway muscles) or other treatment can be provided.
Numerous types of respiratory sensors are suitable for use in the practice of the invention. For example, the implantable sensor may be a respiration element implanted in the thoracic cavity which is capable of generating a respiration signal as part of a ventilation system for providing gas to a host. See, e.g., U.S. Patent No. 6,357,438. The implantable sensor may be composed of a sensing element connected to a lead body which is inserted into bone (e.g., manubrium) that communicates with the intrathoracic cavity to detect respiratory changes. See, e.g., U.S. Patent No. 6,572,543.
Regardless of the specific design features of the respiratory sensor, for accurate detection of physical and/or physiological properties, the device must be accurately positioned adjacent to the tissue. If excessive scar tissue growth or extracellular matrix deposition occurs around the pulmonary function or airway sensing device, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of biological information to the detector mechanism of the sensor. For example, many respiratory sensors (pulmonary function sensing devices) fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (i.e., the device detects conditions in the microenvironment of the capsule surrounding the implant, not the functioning of the respiratory system as whole). Respiratory sensor devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. Respiratory sensing devices having the subject compositions infiltrated into tissue adjacent to the implant can increase the efficiency of detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the device is or will be implanted. In another aspect, the present invention provides respiratory sensor devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with respiratory sensor devices have been described above. Agents or compositions of the present invention may be infiltrated around implanted respiratory sensor devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the respiratory sensor device; (b) the vicinity of the respiratory sensor device-tissue interface; (c) the region around the respiratory sensor device; and (d) tissue surrounding the respiratory sensor device. Methods for infiltrating the subject compositions into tissue adjacent to a respiratory sensor device include delivering the composition: (a) to the surface of the respiratory sensor device (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the respiratory sensor device; (c) to the surface of the respiratory sensor device and/or the tissue surrounding the implanted respiratory sensor device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the respiratory sensor device; (d) by topical application of the composition into the anatomical space where the respiratory sensor device may be placed
(particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the respiratory sensor device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, sensor only, detector only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to respiratory sensor devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced. Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As respiratory sensor devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti- scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2. Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8 - 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10'7, or about 10'7 to 10'6 about 10'6 to 10'5 or about 10'5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
(5) Auditory Sensors
In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to an auditory sensor device. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
The implantable sensor may be a device configured to detect properties in the auditory system. Auditory sensors are used as part of implantable hearing systems for rehabilitation of pure sensorineural hearing losses, or combined conduction and inner ear hearing impairments. Hearing systems may include an implantable sensor which delivers an electrical signal which is processed by an implanted processor and delivered to an implantable electromechanical transducer which acts on the middle or inner ear. The auditory sensor acts as the microphone of the hearing system and acts to convert the incident airborne sound into an electrical signal.
Numerous types of auditory sensors as part of a hearing system are suitable for use in the practice of the invention. For example, the implantable sensor may generate an electrical audio signal as part of a hearing system for rehabilitation of hearing loss. See, e.g., U.S. Patent No. 6,334,072. The implantable sensor may be a capacitive sensor which is mechanically or magnetically coupled to a vibrating auditory element, such as the malleus, which detects the time-varying capacitance values resulting from the vibrations. See, e.g., U.S. Patent No. 6,190,306. The implantable sensor may be an electromagnetic sensor having a permanent magnet and a coil and a time- varying magnetic flux linkage based on the vibrations which are provided to an output stimulator for mechanical or electrical stimulation of the cochlea. See, e.g., U.S. Patent No. 5,993,376.
Auditory sensors, which may benefit from having the subject composition infiltrated into adjacent tissue according to the present invention, include commercially available products. Commercially available auditory sensor devices suitable for the practice of the invention include: the HIRES 9OK Bionic Ear Implant, HIRESOLUTION SOUND, CLARION CII Bionic Ear, and CLARION 1.2, from Advanced Bionics (Sylmar, California, a Boston Scientific Company, see advancedbionics.com); see also U.S. Patent Nos. 6,778,858; 6,754,537; 6,735,474; 6,731,986; 6,658,302; 6,636,768; 6,631,296; 6,628,991 ; 6,498,954; 6,487,453; 6,473,651 ; 6,415,187; and 6,415,185; the NUCLEUS 3 cochlear implant from Cochlear (Lane Cove NSW, Australia, see cochlear.com); see also U.S. Patent Nos. 6,810,289; 6,807,455; 6,788,790; 6,782,619; 6,751 ,505; 6,736,770; 6,700,982; 6,697,674; 6,678,564; 6,620,093; 6,575,894; 6,570,363; 6,565,503; 6,554,762; 6,537,200; 6,525,512; 6,496,734; 6,480,820; 6,421 ,569; 6,411 ,855; 6,394,947; 6,392,386; 6,377,075; 6,301 ,505; 6,289,246; 6,116,413; 5,720,099; 5,653,742; 5,645,585; and U.S. Patent Application Publication Nos. 2004/0172102A1 and 2002/0138115A1 ; the PULSAR Cl 100 and COMBI 40+ cochlear implants from Med-EI (Austria, see medel.com); see also US Patent Application 20040039245A1 , US Patent Nos. 6,600,955; 6,594,525; 6,556,870; and 5,983,139; the ALLHEAR implants from AIIHear, Inc. (Aurora, Oregon; see allhear.com); see also WO 01/50816; EP 1 245 134; and the DIGISONIC CONVEX, DIGISONIC AUDITORY BRAINSTEM, and DIGISONIC MULTI-ARRAY implants from MXM (France; see mxmlab.com); see also U.S. Patent Nos. 5,123,422; EP 0 219 380; WO 04/002193; EP 1 244400 A1 ; US 6,428,484; US 20020095194A1; WO 01/50992.
Regardless of the specific design features of the auditory sensor, for accurate detection of sound, the device must be accurately positioned within the ear. If excessive scar tissue growth or extracellular matrix deposition occurs around the auditory sensor, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of sound waves to the detector mechanism of the sensor. Auditory sensor devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. Auditory sensing devices having the subject compositions infiltrated into tissue adjacent to the implant can increase the efficiency of sound detection and increase the duration that these devices function clinically. In one aspect, the device includes implantable sensor devices having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the device is or will be implanted. In another aspect, the present invention provides auditory sensor devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with auditory sensor devices have been described above.
Agents or compositions of the present invention may be infiltrated around implanted auditory sensor devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the auditory sensor device; (b) the vicinity of the auditory sensor device-tissue interface; (c) the region around the auditory sensor device; and (d) tissue surrounding the auditory sensor device. Methods for infiltrating the subject compositions into tissue adjacent to an auditory sensor device include delivering the composition: (a) to the surface of the auditory sensor device (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue
(e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the auditory sensor device; (c) to the surface of the auditory sensor device and/or the tissue surrounding the implanted auditory sensor device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the auditory sensor device; (d) by topical application of the composition into the anatomical space where the auditory sensor device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the auditory sensor device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, sensor only, detector only and/or a combination thereof. According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to auditory sensor devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As auditory sensor devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8 - 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10"8 - 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should to be maintained on the implant or barrier surface. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1 % of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10"5 or about 10'5 to 10'4 of the agent is maintained on the tissue surface. it should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
(6) Electrolyte and Metabolite Sensors In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to an electrolyte and/or metabolite sensor device. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
In another aspect, implantable sensors may be used to detect electrolytes and metabolites in the blood. For example, the implantable sensor may be a device to monitor constituent levels of metabolites or electrolytes in the blood by emitting a source of radiation directed towards blood such that it interacts with a plurality of detectors that provide an output signal. See, e.g., U.S. Patent No. 6,122,536. The implantable sensor may be a biosensing transponder which is composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader. See, e.g., U.S. Patent No. 5,833,603. The implantable sensor may be a monolithic bioelectronic device for detecting at least one analyte within the body of an animal. See, e.g., U.S. Patent No. 6,673,596. Other sensors that measure chemical analytes are described in, e.g., U.S. Patent Nos. 6,625,479 and 6,201 ,980. If excessive scar tissue growth or extracellular matrix deposition occurs around the sensor, the sensor may receive erroneous information that compromises its efficacy, or the scar tissue may block the flow of metabolites or electrolytes to the detector mechanism of the sensor. For example, many metabolite/electrolyte sensing devices fail after initially successful implantation because encapsulation of the implant causes it to detect nonrelevant levels (i.e., the device detects conditions in the microenvironment of the capsule surrounding the implant, not blood levels). Sensing devices having the subject compositions infiltrated into tissue adjacent to the implant can increase the efficiency of metabolite/electrolyte detection and increase the duration that these devices function clinically. Electrolyte and/or metabolite sensor device such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. In one aspect, the device includes implantable metabolite/electrolyte sensor devices having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the device is or will be implanted. In another aspect, the present invention provides metabolite/electrolyte sensor devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with metabolite/electrolyte sensor devices have been described above.
Agents or compositions of the present invention may be infiltrated around implanted metabolite/electrolyte sensor devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the metabolite/electrolyte sensor device; (b) the vicinity of the metabolite/electrolyte sensor device-tissue interface; (c) the region around the metabolite/electrolyte sensor device; and (d) tissue surrounding the metabolite/electrolyte sensor device. Methods for infiltrating the subject compositions into tissue adjacent to a metabolite/electrolyte sensor device include delivering the composition: (a) to the surface of the metabolite/electrolyte sensor device (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the metabolite/electrolyte sensor device; (c) to the surface of the metabolite/electrolyte sensor device and/or the tissue surrounding the implanted metabolite/electrolyte sensor device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the metabolite/electrolyte sensor device; (d) by topical application of the composition into the anatomical space where the metabolite/electrolyte sensor device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the metabolite/electrolyte sensor device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, sensor only, detector only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to metabolite/electrolyte sensor devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As metabolite/electrolyte sensor devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10~4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10~7, or about 10'7 to 10'6 about 10"6 to 10"5 or about 10~5 to 10'4 of the agent is maintained on the tissue surface. It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouraciI), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Although numerous examples of implantable sensor devices have been described above, all possess similar design features and cause similar unwanted foreign body tissue reactions following implantation and may introduce or promote infection in the area of the implant site. It should be obvious to one of skill in the art that commercial sensor devices not specifically cited above as well as next-generation and/or subsequently developed commercial sensor products are to be anticipated and are suitable for use under the present invention. The sensor device, particularly the sensing element, must be positioned in a very precise manner to ensure that detection is carried out at the correct anatomical location in the body. All, or parts, of a sensor device can migrate following surgery, or excessive scar tissue growth can occur around the implant, which can lead to a reduction in the performance of these devices. The formation of a fibrous capsule around the sensor can impede the flow of biological information to the detector and/or cause the device to detect levels that are not physiologically relevant (Ae., detect levels in the capsule instead of true physiological levels outside the capsule). Not only can this lead to incomplete or inaccurate readings, it can cause the physician or the patient to make incorrect therapeutic decisions based on the information generated. Implantable sensor devices having the subject compositions infiltrated into tissue adjacent to the sensor-tissue interface can be used to increase the efficacy and/or the duration of activity of the implant. Implantable sensor devices may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. In one aspect, the present invention provides implantable sensor devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). These compositions can further include one or more fibrosis-inhibiting agents such that the overgrowth of granulation, fibrous, or neointimal tissue is inhibited or reduced and/or one or more anti-infective agents such that infection in the vicinity of the implant site is inhibited or prevented.
Implantable Drug Delivery Devices and Pumps
In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to an implantable drug delivery device or pump. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Implantable drug delivery devices and pumps are a means to provide prolonged, site-specific release of a therapeutic agent for the management of a variety of medical conditions. Drug delivery implants and pumps are generally utilized when a localized pharmaceutical impact is desired (Ae., the condition affects only a specific region) or when systemic delivery of the agent is inefficient or ineffective (Ae., leads to toxicity or severe side effects, results in inactivation of the drug prior to reaching the target tissue, produces poor symptom/disease control, and/or leads to addiction to the medication). Implantable pumps can also deliver systemic drug levels in a constant, regulated manner for extended periods and help patients avoid the "peaks and valleys" of blood-level drug concentrations associated with intermittent systemic dosing. Another advantage of implantable pumps is improved patient compliance. Many patients forget to take their medications regularly (particularly the young, elderly, chronically ill, mentally handicapped), but with an implantable pump, this problem is alleviated. For many patients this can lead to better symptom control (the dosage can often be titrated to the severity of the symptoms), superior disease management (particularly for insulin delivery in diabetics), and lower drug requirements (particularly for pain medications).
Innumerable drug delivery implants and pumps have been used in a variety of clinical applications, including programmable insulin pumps for the treatment of diabetes, intrathecal (in the spine) pumps to administer narcotics (e.g., morphine, fentanyl) for the relief of pain (e.g., cancer, back problems, HIV, post-surgery), local and systemic delivery of chemotherapy for the treatment of cancer (e.g., hepatic artery 5-FU infusion for liver tumors), medications for the treatment of cardiac conditions (e.g. , anti-arrhythmic drugs for cardiac rhythm abnormalities), intrathecal delivery of anti-spasmotic drugs (e.g., baclofen) for spasticity in neurological disorders (e.g., Multiple Sclerosis, spinal cord injuries, brain injury, cerebral palsy), or local/regional antibiotics for infection management (e.g., osteomyelitis, septic arthritis). Typically, drug delivery pumps are implanted subcutaneously and consist of a pump unit with a drug reservoir and a flexible catheter through which the drug is delivered to the target tissue. The pump stores and releases prescribed amounts of medication via the catheter to achieve therapeutic drug levels either locally or systemically (depending upon the application). The center of the pump has a self-sealing access port covered by a septum such that a needle can be inserted percutaneously (through both the skin and the septum) to refill the pump with medication as required. There are generally two types of implantable drug delivery pumps. Constant-rate pumps are usually powered by gas and are designed to dispense drugs under pressure as a continual dosage at a preprogrammed, constant rate. The amount and rate of drug flow and regulated by the length of the catheter used, temperature, and altitude and they are best when unchanging, long-term drug delivery is required. Programmable- rate pumps utilize a battery-powered pump and a constant pressure reservoir to deliver drugs on a periodic basis in a manner that can be programmed by the physician or the patient. For the programmable infusion device, the drug may be delivered in small, discrete doses based on a programmed regimen which can be altered according to an individual's clinical response.
In general, drug delivery pumps are implanted to deliver drug at a regulated dose and may, in certain applications, be used in conjunction with implantable sensors that collect information which is used to regulate drug delivery (often called a "closed loop" system). Implantable drug delivery pumps may function and deliver drug in a variety of ways, which include, but are not limited to: (a) delivering drugs only when changes in the body are detected (e.g., sensor stimulated); (b) delivering drugs as a continuous slow release (e.g., constant flow); (c) delivering drugs at prescribed dosages in a pulsatile manner (e.g., non-constant flow); (d) delivering drugs by programmable means; and (e) delivering drugs through a device that is designed for a specific anatomical site (e.g., intraocular, intrathecal, intraperitoneal, intra-arterial or intracardiac). In addition to delivering drugs in a specific way or to a specific location, drug delivery pumps may also be categorized based on their mechanical delivery technology (e.g., the driving force by which drug delivery occurs). For example, the mechanics for delivering drugs may include, without limitation, osmotic pumps, metering systems, peristaltic (roller) pumps, electronically driven pumps, ocular drug delivery pumps and implants, elastomeric pumps, spring-contraction pumps, gas-driven pumps (e.g., induced by electrolytic cell or chemical reaction), hydraulic pumps, piston-dependent pumps and non-piston-dependent pumps, dispensing chambers, infusion pumps, passive pumps, infusate pumps and osmotically-driven fluid dispensers.
The clinical function of an implantable drug delivery device or pump depends upon the device, particularly the catheter or drug-dispensing component(s), being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid) and not becoming encapsulated or obstructed by scar tissue. Unfortunately, in many instances when these devices are implanted in the body, they are subject to a "foreign body" response from the surrounding host tissues as described previously. For implantable pumps, the drug-delivery catheter lumen, catheter tip, dispensing components, or delivery membrane may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Alternatively, the entire pump, the catheter and/or the dispensing components can become encapsulated by scar (i.e., the body "walls off' the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule). Either of these developments may lead to inefficient or incomplete drug flow to the desired target tissues or organs (and loss of clinical benefit), while encapsulation can also lead to local drug accumulation (in the capsule) and additional clinical complications (e.g., local drug toxicity; drug sequestration followed by sudden "dumping" of large amounts of drug into the surrounding tissues). Additionally, the tissue surrounding the implantable pump can be inadvertently damaged from the inflammatory foreign body response leading to loss of function and/or tissue damage (e.g., scar tissue in the spinal canal causing pain or obstructing the flow of cerebrospinal fluid). Implantation of an implantable drug delivery device or pump may also introduce or promote infection in the vicinity of the implant site. Implantable drug delivery pumps that release one or more therapeutic agents for reducing scarring at the device-tissue interface (particularly in and around the drug delivery catheter or drug dispensing components) may help prolong the clinical performance of these devices. Inhibition of fibrosis can make sure that the correct amount of drug is dispensed from the device at the appropriate rate and that potentially toxic drugs do not become sequestered in a fibrous capsule. For devices that include electrical or battery components, not only can fibrosis cause the device to function suboptimally or not at all, it can cause excessive drain on battery life as increased energy is required to overcome the increased resistance imposed by the intervening scar tissue. Implantation of an implantable drug delivery device or pump may also introduce or promote infection in the vicinity of the implant site.
Virtually any implantable pump may benefit from the present invention. In one aspect, the drug delivery pump may deliver drugs in a continuous, constant-flow, slow release manner. For example, the drug delivery pump may be a passive pump adapted to provide a constant flow of medication which may be regulated by a pressure sensing chamber and a valve chamber in which the constant flow rate may be changed to a new constant flow rate. See, e.g., U.S. Patent No. 6,589,205. In another aspect, the drug delivery pump may deliver drugs at prescribed dosages in a non-constant flow or pulsatile manner. For example, the drug delivery pump may adapt a regular pump to generate a pulsatile fluid drug flow by continuously filling a chamber and then releasing a valve to provide a bolus pulse of the drug. See, e.g., U.S. Patent No. 6,312,409. In another aspect, the drug delivery pump may be programmed to dispense drug in a very specific manner. For example, the drug delivery pump may be a programmable infusate pump composed of a variable volume infusate chamber, and variable volume control fluid pressure and displacement reservoirs, whereby a fluid flow is sampled by a microprocessor based on the programmed value and adjustments are made accordingly to maintain the programmed fluid flow. See, e.g., U.S. Patent No. 4,443,218. In another aspect, the drug delivery pump suitable for use in the present invention may be manufactured based on different mechanical technologies (e.g., driving forces) of delivering drugs. For example, the drug delivery pump may be an implant composed of a piston that divides two chambers in which one chamber contains a water-swellable agent and the other chamber contains a leuprolide formulation for delivery. See, e.g., U.S. Patent No. 5,728,396. The drug delivery pump may be a non-cylindrical osmotic pump system that may not rely upon a piston to infuse drug and conforms to the anatomical implant site. See, e.g., U.S. Patent No. 6,464,688. The drug delivery pump may be an osmotically driven fluid dispenser composed of a flexible inner bag that contains the drug composition and a port in which the composition can be delivered. See, e.g., U.S. Patent No. 3,987,790. The drug delivery pump may be a fluid-imbibing delivery implant composed of a compartment with a composition permeable to the passage of fluid and has an extended rigid sleeve to resist transient mechanical forces. See, e.g., U.S. Patent Nos. 5,234,692 and 5,234,693. The drug delivery pump may be a pump with an isolated hydraulic reservoir, metering device, displacement reservoir, drug reservoir, and drug infusion port that is all contained in a housing apparatus. See, e.g., U.S. Patent No. 6,629,954. The drug delivery pump may be composed of a dispensing chamber that has a dispensing passage and valves that are under compressive force to enable drug to flow in a one-way direction. See, e.g., U.S. Patent No. 6,283,949. The drug delivery pump may be spring-driven based on a spring regulating pressure difference with a variable volume drug chamber. See, e.g., U.S. Patent No. 4,772,263. Other examples of drug delivery pumps are described in, e.g., U.S. Patent Nos. 6,645,176; 6,471 ,688; 6,283,949; 5,137,727 and 5,112,614. Implantable drug delivery devices and pumps, which may benefit from having the subject composition infiltrated into adjacent tissue according to the present invention, include commercially available products. For example, there are osmotically driven drug delivery pumps that are commercially available and suitable for the practice of the invention. These osmotic pumps include the DUROS Implant and ALZET Osmotic Pump from Alza Corporation (Mountain View, CA), which are used to delivery a wide variety of drugs and other therapeutics through the method of osmosis (see, e.g., U.S. Patent Nos. 6,283,953; 6,270,787; 5,660,847; 5,112,614; 5,030,216 and 4,976,966).
As described above, infiltration of the subject composition into tissue adjacent to the drug delivery pump can improve performance of the device and/or prevent or inhibit infection in the vicinity of the implant site. In one aspect, the present invention provides implantable drug delivery devices and pumps having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti- scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with implantable drug delivery devices and pumps have been described above.
Agents or compositions of the present invention may be infiltrated around implanted implantable drug delivery devices and pumps by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the implantable drug delivery device or pump; (b) the vicinity of the implantable drug delivery device or pump-tissue interface; (c) the region around the implantable drug delivery device or pump; and (d) tissue surrounding the implantable drug delivery device or pump. Methods for infiltrating the subject compositions into tissue adjacent to an implantable drug delivery device or pump include delivering the composition: (a) to the surface of the implantable drug delivery device or pump (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the implantable drug delivery device or pump; (c) to the surface of the implantable drug delivery device or pump and/or the tissue surrounding the implanted implantable drug delivery device or pump (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the implantable drug delivery device or pump; (d) by topical application of the composition into the anatomical space where the implantable drug delivery device or pump may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the implantable drug delivery device or pump as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, pump only, catheter only, drug dispensing components only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to implantable drug delivery devices and pumps may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced. Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As implantable drug delivery devices and pumps are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti- scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 -40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC5O range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8 - 10'4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10'8 to 10'7, or about 10"7 to 10"6 about 10"6 to 10"5 or about 10"5 to 10'4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
It should be obvious to one of skill in the art that commercial drug delivery pumps not specifically cited as well as next-generation and/or subsequently-developed commercial drug delivery products are to be anticipated and are suitable for use under the present invention.
Several specific drug delivery pumps and treatments will be described in greater detail below.
(1) Implantable Insulin Pumps for Diabetes In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to an insulin pump. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti- scarring and/or anti-infective agent).
Insulin pumps are used for patients with diabetes to replace the need to control blood glucose levels by daily manual injections of insulin. Precise titration of the dosage and timing of insulin administration is a critical component in the effective management of diabetes. If the insulin dosage is too high, blood glucose levels drop precipitously, resulting in confusion and potentially even loss of consciousness. If insulin dosage is too low, blood glucose levels rise too high, leading to excessive thirst, urination, and changes in metabolism known as ketoacidosis. If the timing of insulin administration is incorrect, blood glucose levels can fluctuate wildly between the two extremes - a situation that is thought to contribute to some of the long-term complications of diabetes such as heart disease, kidney failure, nerve damage and blindness. Since in the extreme, all these conditions can be life threatening, the precise dosing and timing of insulin administration is essential to preventing the short and long-term complications of diabetes.
Implantable pumps automate the administration of insulin and eliminate human errors of dosage and timing that can have long-term health consequences. The pump has the capability to inject insulin regularly, multiple times a day and in small doses into the blood stream, peritoneal cavity or subcutaneous tissue. The pump is refilled with insulin once or twice a month by injection directly into the pump chamber. This reduces the number of externally administered injections the patient must undergo and also allows preprogrammed variable amounts of insulin to be released at different times into the blood stream; a situation which more closely resembles normal pancreas function and minimizes fluctuations in blood glucose levels. The insulin pump may be activated by an externally generated signal after the patient has withdrawn a drop of blood, subjected it to an analysis, and made a determination of the amount of insulin that needs to be delivered. However, the most widely pursued application of this technology is the production of a closed- loop "artificial pancreas" which can continuously detect blood glucose levels (through an implanted sensor) and provide feedback to an implantable pump to modulate the administration of insulin to a diabetic patient.
Numerous types of insulin pumps are suitable for use in the practice of the invention. For example, the drug delivery pump may include both an implantable sensor and a drug delivery pump by being composed of a mass of living cells and an electrical signal that regulates the delivery of glucose or glucagon or insulin. See, e.g., U.S. Patent No. 5,474,552. The drug delivery pump may be composed of a single channel catheter with a sensor which is implanted in a vessel that transmits blood chemistry to a subcutaneously implanted infusion device which then dispenses medication through the catheter. See, e.g., U.S. Patent No. 5,109,850.
Insulin pumps, which may benefit from having the subject composition infiltrated into adjacent tissue according to the present invention, include commercially available products. Commercially available insulin pump devices suitable for the practice of the invention include the MINIMED 2007 Implantable Insulin Pump System from Medtronic MiniMed, Inc. (Northridge, CA). The MINIMED pump delivers insulin into the peritoneal cavity in short, frequent bursts to provide insulin to the body similar to that of the normal pancreas (see, e.g., U.S. Patent Nos. 6,558,345 and 6,461,331). The
MINIMED 2001 Implantable Insulin Pump System (Medtronic MiniMed Inc., Northridge, CA) delivers intraperitoneal insulin injections in a pulsatile manner from a negative pressure reservoir. Both these devices feature a long catheter that transports insulin from the subcutaneously implanted pump into the peritoneal cavity. As described above, the peritoneal drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Insulin pump devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the catheter and/or vicinity of the implant site. In one aspect of the present invention, the device includes delivery catheters having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the delivery catheter is or will be implanted to keep the delivery catheter lumen patent and/or prevent fibrosis in the surrounding tissue and/or inhibit or prevent infection in the catheter or vicinity of the implant site. In another aspect, the present invention provides insulin pumps having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with insulin pumps have been described above. Agents or compositions of the present invention may be infiltrated around implanted insulin pumps by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the insulin pump; (b) the vicinity of the insulin pump-tissue interface; (c) the region around the insulin pump; and (d) tissue surrounding the insulin pump. Methods for infiltrating the subject compositions into tissue adjacent to a insulin pump include delivering the composition: (a) to the surface of the insulin pump (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the insulin pump; (c) to the surface of the insulin pump and/or the tissue surrounding the implanted insulin pump (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the insulin pump; (d) by topical application of the composition into the anatomical space where the insulin pump may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the insulin pump as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, pump only, catheter only, drug dispensing components only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to insulin pumps may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As insulin pumps are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10"8 - 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1 % of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10"5 or about 10"5 to 10'4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
It should be obvious to one of skill in the art that commercial drug delivery pumps not specifically cited as well as next-generation and/or subsequently-developed commercial drug delivery products are to be anticipated and are suitable for use under the present invention. (2) Intrathecal Drug Delivery Pumps In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to an intrathecal drug delivery pump. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Intrathecal drug delivery pumps having the subject composition infiltrated into tissue adjacent to the pump may used to deliver drugs into the spinal cord for pain management and movement disorders.
Chronic pain is one of the most important clinical problems in all of medicine. For example, it is estimated that over 5 million people in the United States are disabled by back pain. The economic cost of chronic back pain is enormous, resulting in over 100 million lost work days annually at an estimated cost of $50-100 billion. The cost of managing pain for oncology patients is thought to approach $12 billion. Chronic pain disables more people than cancer or heart disease and costs the American public more than both cancer and heart disease combined. In addition to the physical consequences, chronic pain has numerous other costs including loss of employment, marital discord, depression and prescription drug addiction. It goes without saying, therefore, that reducing the morbidity and costs associated with persistent pain remains a significant challenge for the healthcare system.
Intractable severe pain resulting from injury, illness, scoliosis, spinal disc degeneration, spinal cord injury, malignancy, arachnoiditis, chronic disease, pain syndromes (e.g., failed back syndrome, complex regional pain syndrome) and other causes is a debilitating and common medical problem. In many patients, the continued use of analgesics, particularly drugs like narcotics, are not a viable solution due to tolerance, loss of effectiveness, and addiction potential. In an effort to combat this, intrathecal drug delivery devices have been developed to treat severe intractable back pain that is resistant to other traditional treatment modalities such as drug therapy, invasive therapy (surgery), or behavioral/lifestyle changes. Intrathecal drug delivery pumps are designed and used to reduce pain by delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord. Typically, since this therapy delivers pain medication topically to pain receptors contained in the spinal cord that transmit pain sensation directly to the brain, smaller doses of medication are needed to gain relief. Morphine and other narcotics (usually fentanyl and sufentanil) are the most commonly delivered agents and many patients receive superior relief with lower doses than can be achieved with systemic delivery. Intrathecal drug delivery also allows the administration of pain medications (such as Ziconotide; an N-type calcium channel blocker made by Elan
Pharmaceuticals) that cannot cross the blood-brain barrier and are thus only effective when administered by this route.
Intrathecal pumps are also used in the management of neurological and movement disorders. Baclofen (marketed as Lioresal by Novartis) is an antispasmotic/muscle relaxant used to treat spasticity and improve mobility in patients with Multiple Sclerosis, cystic fibrosis and spinal injuries. This drug has been proven to be more effective and cause fewer side effects when administered into the CSF by an intrathecal drug delivery pump. Efforts are also underway to treat epilepsy, brain tumors, Alzheimer's disease, Parkinson's disease and Amyetropic Lateral Sclerosis (ALS - Lou Gehrig's disease) via intrathecal administration of agents that may be too toxic to deliver systemically or do not cross the blood-brain barrier. For example, trials of intrathecal^ administered recombinant brain-derived neurotrophic factor (r- BDNF made by Amgen) have been undertaken in ALS patients. An intrathecal drug delivery system consists of an intrathecal drug infusion pump and an intraspinal catheter, both of which are fully implanted. The pump device is implanted under the skin in the abdominal area, just above or below the beltline and can be refilled by percutaneous injection of the drug into the reservoir. The catheter is tunneled under the skin and runs from the pump to the intrathecal space of the spine. When operational, the pump administers prescribed amounts of medication to the cerebrospinal fluid in either a continuous fashion or in a manner than can be controlled by the physician or the patient in response to symptoms.
Numerous types of implantable intrathecal pumps are suitable for use in the practice of the invention. For example, the implantable pump used to deliver medication may be composed of two osmotic pumps with semipermeable membranes configured to deliver up to two drug delivery regimens at different rates, and having a built-in backup drug delivery system whereby the delivery of drug may continue when the primary delivery system reaches the end of its useful life or fails unexpectedly. See, e.g., U.S. Patent No. 6,471,688. The implantable pump may be may be composed of a battery- operated pump unit with a drug reservoir, catheter, and electrodes that are implanted in the epidural space of a patient for relief of pain by delivering a liquid pain-relieving agent through the catheter to the desired location. See, e.g., U.S. Patent No. 5,458,631. Similar drug-delivery pumps have been described for the infusion of agents into regions of the brain to locally affect the excitability of the neurons in the treatment of a variety of chronic neurogenerative diseases (such as those described above for intrathecal delivery). Implantable pumps may be implanted abdominally which then dispenses drug through a catheter that is tunneled from the abdominal implant site, through the neck to an entry site in the head, and then to the localized treatment site within the brain. Pumps that deliver drug to the brain may discharge the drug at a variety of locations, including, but not limited to, anterior thalamus, ventrolateral thalamus, internal segment of the globus pallidus, substantia nigra pars reticulate, subthalamic nucleus, external segment of globus pallidus, and neostriatum. For example, the drug delivery pump may be composed of an implantable pump portion coupled to a catheter for infusing dosages of drug to a predetermined location of the brain when a sensor detects a symptom, such that a neurological disorder (e.g., seizure) may be treated. See, e.g., U.S. Patent No. 5,978,702. The implantable pump may be implanted adjacent to a predetermined infusion site in a brain such that a predetermined dosage of at least one drug capable of altering the level of excitation of neurons of the brain may be infused such that neurodegeneration is prevented and/or treated. See, e.g., U.S. Patent No. 5,735,814. The implantable pump may include a reservoir for the therapeutic agent which is stored between the galea aponeurotica and cranium of a subject whereby drug is then dispensed via pumping action to the desired location. See, e.g., U.S. Patent No. 6,726,678.
Intrathecal drug delivery pumps, which may benefit from having the subject composition infiltrated into adjacent tissue according to the present invention, include commercially available products. There are numerous commercially available implantable, intrathecal drug-delivery systems which are suitable for the practice of the invention. The SYNCHROMED EL Infusion System which is made by Medtronic, Inc. and is indicated for chronic Intrathecal Baclofen Therapy (ITB Therapy) (see, e.g., U.S. Patent Nos. 6,743,204; 6,669,663; 6,635,048; 6,629,954; 6,626,867; 6,102,678; 5,978,702 and 5,820,589) The SYNCHROMED pump is a programmable, battery-operated device that stores and delivers medication based on the programmed dosing regimen. Medtronic, Inc. (Minneapolis, MN) also sells their ISOMED Constant- Flow Infusion System for use in delivering morphine sulfate directly into the intrathecal space as a treatment for chronic pain. Arrow International produces the Model 3000 infusion pump that provides constant-rate administration of agents such as morphine and baclofen into the intrathecal space. Tricumed Medizintechnik GmbH (Kiel, Germany) produces the Archimedes® constant flow implantable infusion pump for intrathecal administration of pain and antispasmotic drugs. Advanced Neuromodulation Systems (Piano, TX) produces the AccuRx® infusion pump for the treatment of pain and neuromuscular disorders. All these devices feature a long catheter that transports the active agent from a subcutaneously implanted pump into the intrathecal space in the spinal cord. As described above, the intrathecal drug- delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Another potential complication with intrathecal drug delivery is the formation of fibrous tissue in the subdural space that can obstruct CSF flow and lead to serious complications (e.g., hydrocephalus, increased intracranial pressure). Intrathecal drug delivery devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the catheter and/or vicinity of the implant site. In one aspect of the present invention, the device includes delivery catheters having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the delivery catheter is or will be implanted to keep the delivery catheter lumen patent and/or prevent fibrosis in the surrounding tissue and/or inhibit or prevent infection in the catheter or vicinity of the implant site. In another aspect, the present invention provides intrathecal drug delivery devices having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with intrathecal drug delivery devices have been described above.
Agents or compositions of the present invention may be infiltrated around implanted intrathecal drug delivery devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the intrathecal drug delivery device; (b) the vicinity of the intrathecal drug delivery device- tissue interface; (c) the region around the intrathecal drug delivery device; and (d) tissue surrounding the intrathecal drug delivery device. Methods for infiltrating the subject compositions into tissue adjacent to an intrathecal drug delivery device include delivering the composition: (a) to the surface of the intrathecal drug delivery device (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the intrathecal drug delivery device; (c) to the surface of the intrathecal drug delivery device and/or the tissue surrounding the implanted intrathecal drug delivery device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the intrathecal drug delivery device; (d) by topical application of the composition into the anatomical space where the intrathecal drug delivery device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the intrathecal drug delivery device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, pump only, catheter only, drug dispensing components only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to intrathecal drug delivery devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As intrathecal drug delivery devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti- scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10'4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^ - 10"4 M of agent should to be maintained on the implant or barrier surface. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G). platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10"5 or about 10'5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
It should be obvious to one of skill in the art that commercial intrathecal drug delivery pumps not specifically cited as well as next-generation and/or subsequently developed commercial drug delivery products are to be anticipated and are suitable for use under the present invention.
(3) Implantable Drug Delivery Pumps for Chemotherapy In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to a chemotherapeutic drug delivery pump. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
The drug delivery pump may be a pump that dispenses a chemotherapeutic drug for the treatment of cancer. Pumps for dispensing a drug for the treatment of cancer are used to deliver chemotherapeutic agents to a local area of the body. Although virtually any malignancy may potentially be treated in this manner (Ae., by infusing drug directly into a solid tumor or into the blood vessels that supply the tumor), current treatments revolve around the management of hepatic (liver) tumors. For example, FUDR (2'-deoxy 5- fluorouridine) is used in the palliative management of adenocarcinoma (colon, breast, stomach) that has metastasized to the liver. In hepatic artery infusion therapy the drug is delivered via an implantable pump into the artery which provides blood supply to the liver. This allows for higher drug concentrations to reach the liver (the drug is not diluted in the blood as may occur in intravenous administration) and prevents clearance by the liver (the drug is metabolized by the liver and may be rapidly cleared from the bloodstream if administered i.v.); both of which allow higher concentrations of the drug to reach the tumor.
Numerous types of implantable pumps are suitable for delivering chemotherapeutic agents in the practice of the invention. For example, the implantable pump may have a dispensing chamber with a dispensing passage and actuator, reservoir housing with reservoir, and septum for refilling the reservoir. See, e.g., U.S. Patent No. 6,283,949. Medtronic, Inc. sells their ISOMED Constant-Flow Infusion System which may be used to deliver chronic intravascular infusion of floxuridine in a fixed flow rate for the treatment of primary or metastatic cancer. Tricumed Medizintechnik GmbH (Kiel, Germany) sells their ARCHIMEDES DC implantable infusion pump specially adapted to deliver chemotherapy in a constant flow rate within the vicinity of a tumor (see, e.g., U.S. Patent Nos. 5,908,414 and 5,769,823). Arrow International produces the Model 3000 infusion pump that provides constant-rate administration of chemotherapeutic agents into a tumor. All these devices feature a catheter that transports the chemotherapeutic agent from a subcutaneously implanted pump directly into the tumor or the artery that supplies a tumor. As described above, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. If placed intravascularly, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by neointimal tissue which may impair the flow of drug into the blood vessel. Chemotherapeutic drug delivery pumps such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the catheter and/or vicinity of the implant site. In one aspect of the present invention, the device includes delivery catheters having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the delivery catheter is or will be implanted to keep the delivery catheter lumen patent and/or prevent fibrosis in the surrounding tissue and/or inhibit or prevent infection in the catheter or vicinity of the implant site. In another aspect, the present invention provides chemotherapeutic drug delivery pumps having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with chemotherapeutic drug delivery pumps have been described above.
Agents or compositions of the present invention may be infiltrated around implanted chemotherapeutic drug delivery pumps by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the chemotherapeutic drug delivery pump; (b) the vicinity of the chemotherapeutic drug delivery pump-tissue interface; (c) the region around the chemotherapeutic drug delivery pump; and (d) tissue surrounding the chemotherapeutic drug delivery pump. Methods for infiltrating the subject compositions into tissue adjacent to a chemotherapeutic drug delivery pump include delivering the composition: (a) to the surface of the chemotherapeutic drug delivery pump (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the chemotherapeutic drug delivery pump; (c) to the surface of the chemotherapeutic drug delivery pump and/or the tissue surrounding the implanted chemotherapeutic drug delivery pump (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the chemotherapeutic drug delivery pump; (d) by topical application of the composition into the anatomical space where the chemotherapeutic drug delivery pump may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the chemotherapeutic drug delivery pump as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, pump only, catheter only, drug dispensing components only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to chemotherapeutic drug delivery pumps may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As chemotherapeutic drug delivery pumps are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface.
Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10'8 - 10"4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-fnfective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10~7, or about 10"7 to 10'6 about 10"6 to 10"5 or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
It should be obvious to one of skill in the art that commercial chemotherapy delivery pumps and implants not specifically cited as well as next-generation and/or subsequently-developed commercial chemotherapy delivery products are to be anticipated and are suitable for use in the present invention.
(4) Drug Delivery Pumps for the Treatment of Heart Disease In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to a drug delivery pump for the treatment of heart disease. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
The drug delivery pump may be a pump that dispenses a drug for the treatment of heart disease. Pumps for dispensing a drug for the treatment of heart disease may be used to treat conditions including, but not limited to atrial fibrillation and other cardiac rhythm disorders. Atrial fibrillation is a form of heart disease that afflicts millions of people. It is a condition in which the normal coordinated contraction of the heart is disrupted, primarily by abnormal and uncontrolled action of the atria of the heart. Normally, contractions occur in a controlled sequence with the contractions of the other chambers of the heart. When the right atrium fails to contract, contracts out of sequence, or contracts ineffectively, blood flow from the atria to the ventricles is disrupted. Atrial fibrillation can cause weakness, shortness of breath, angina, lightheadedness and other symptoms due to reduced ventricular filling and reduced cardiac output. Stroke can occur as a result of clot forming in a poorly contracting atria, breaking loose, and traveling via the bloodstream to the arteries of the brain where they become wedged and obstruct blood flow (which may lead to brain damage and death). Typically, atrial fibrillation is treated by medical or electrical conversion (defibrillation), however, complications may exist whereby the therapy causes substantial pain or has the potential to initiate a life threatening ventricular arrhythmia. The pain associated with the electrical shock is severe and unacceptable for many patients, since they are conscious and alert when the device delivers electrical therapy. Medical therapy involves the delivery of anti-arrhythmic drugs by injecting them intravenously, administering them orally or delivering them locally via a drug delivery pump. Numerous types of implantable pumps are described for dispensing a drug for the treatment of heart disease and are suitable for use in the practice of the invention. For example, the drug delivery pump may be an implantable cardiac electrode which delivers stimulation energy and dispenses drug adjacent to the stimulation site. See, e.g., U.S. Patent No. 5,496,360. The drug delivery pump may have a plurality of silicone septii to facilitate the filling of drug reservoirs within the pump which is subcutaneously implanted with a catheter which travels transvenously by way of the subclavian vein through the superior vena cava and into the right atrium for drug delivery. See, e.g., U.S. Patent No. 6,296,630. As described above, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. If placed intravascularly, the drug-delivery catheter lumen or catheter tip may become partially or fully obstructed by neointimal tissue which may impair the flow of drug into the blood vessel or the right atrium. Drug delivery pumps such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the catheter and/or vicinity of the implant site. In one aspect of the present invention, the device includes delivery catheters having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the delivery catheter is or will be implanted to keep the delivery catheter lumen patent and/or prevents fibrosis in the surrounding tissue and/or inhibit or prevent infection in the catheter or vicinity of the implant site. In another aspect, the present invention provides drug delivery pumps for the treatment of heart disease having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non-polymeric delivery systems for use in connection with drug delivery pumps for the treatment of heart disease have been described above.
Agents or compositions of the present invention may be infiltrated around implanted drug delivery pumps for the treatment of heart disease by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the drug delivery pump for the treatment of heart disease; (b) the vicinity of the drug delivery pump for the treatment of heart disease-tissue interface; (c) the region around the drug delivery pump for the treatment of heart disease; and (d) tissue surrounding the drug delivery pump for the treatment of heart disease. Methods for infiltrating the subject compositions into tissue adjacent to a drug delivery pump for the treatment of heart disease include delivering the composition: (a) to the surface of the drug delivery pump for the treatment of heart disease (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the drug delivery pump for the treatment of heart disease; (c) to the surface of the drug delivery pump for the treatment of heart disease and/or the tissue surrounding the implanted drug delivery pump for the treatment of heart disease (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the drug delivery pump for the treatment of heart disease; (d) by topical application of the composition into the anatomical space where the drug delivery pump for the treatment of heart disease may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the drug delivery pump for the treatment of heart disease as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, pump only, catheter only, drug dispensing components only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to drug delivery pumps for the treatment of heart disease may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As drug delivery pumps for the treatment of heart disease are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2. Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10~8- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531
(ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10~8- 10~4 M of agent should to be maintained on the implant or barrier surface. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10'8 to 10"7, or about 10'7 to 10"6 about 10"6 to 10"5 or about 10'5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines {e.g., doxorubicin or mitoxantrone), fluoropyrimidines {e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
It should be obvious to one of skill in the art that commercial cardiac drug delivery pumps not specifically cited as well as next-generation and/or subsequently-developed commercial cardiac drug delivery products are to be anticipated and are suitable for use under the present invention.
(5) Other Drug Delivery Implants In one aspect, the subject agents or compositions may be infiltrated into tissue adjacent to an implantable pump for continuous delivery of pharmaceutical agents. The subject compositions (e.g., polymer compositions) may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Several other implantable pumps useful in the present invention have been developed for continuous delivery of pharmaceutical agents. For example, Debiotech SA (Switzerland) has developed the MIP device which is an implantable piezo-actuated silicon micropump for programmable drug delivery applications. This high-performance micropump is based on a MEMS (Micro-Electro-Mechanical) system which allows it to maintain a low flow rate. The DUROS sufentanil implant from Durect Corporation (Cupertino, CA) is a titanium cylinder that contains a drug reservoir, and a piston driven by an osmotic engine. The VIADUR (leuprolide acetate) implant available from Alza Corporation (Mountain View, CA) uses the same DUROS implant technology to deliver leuprolide over a 12 month period to reduces testosterone levels for the treatment prostate cancer (see, e.g., U.S. Patent Nos. 6,283,953; 6,270,787; 5,660,847; 5,112,614; 5,030,216 and 4,976,966). Fibrous encapsulation of the device can cause failure in a number of ways including: obstructing the semipermeable membrane (which will impair functioning of the osmotic engine by preventing the flow of fluids into the engine), obstructing the exit port (which will impair drug flow out of the device) and/or complete encapsulation (which will create a microenvironment that prevents drug distribution). Many other drug delivery implants, osmotic pumps and the like suffer from similar problems - fibrous encapsulation prevents the appropriate release of drugs into the surrounding tissues. Drug delivery devices such as these may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the catheter and/or vicinity of the implant site. In one aspect of the present invention, drug delivery devices having the subject composition comprising an anti-scarring agent and/or anti-infective agent infiltrated into tissue adjacent to where the device is or will be implanted to prevent or inhibit encapsulation, prevent obstruction of the semipermeable membrane, keep the delivery catheter lumen patent, prevent fibrosis in the surrounding tissue and/or inhibit or prevent infection in the catheter or vicinity of the implant site. In one aspect, the present invention provides implantable pumps for continuous delivery of pharmaceutical agents having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Numerous polymeric and non- polymeric delivery systems for use in connection with implantable pumps for continuous delivery of pharmaceutical agents have been described above.
Agents or compositions of the present invention may be infiltrated around implanted implantable pumps for continuous delivery of pharmaceutical agents by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the implantable pump for continuous delivery of pharmaceutical agents; (b) the vicinity of the implantable pump for continuous delivery of pharmaceutical agents-tissue interface; (c) the region around the implantable pump for continuous delivery of pharmaceutical agents; and (d) tissue surrounding the implantable pump for continuous delivery of pharmaceutical agents. Methods for infiltrating the subject compositions into tissue adjacent to an implantable pump for continuous delivery of pharmaceutical agents include delivering the composition: (a) to the surface of the implantable pump for continuous delivery of pharmaceutical agents (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the implantable pump for continuous delivery of pharmaceutical agents; (c) to the surface of the implantable pump for continuous delivery of pharmaceutical agents and/or the tissue surrounding the implanted implantable pump for continuous delivery of pharmaceutical agents (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the implantable pump for continuous delivery of pharmaceutical agents; (d) by topical application of the composition into the anatomical space where the implantable pump for continuous delivery of pharmaceutical agents may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the implantable pump for continuous delivery of pharmaceutical agents as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies [i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device, including the device only, pump only, catheter only, drug dispensing components only and/or a combination thereof.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to implantable pumps for continuous delivery of pharmaceutical agents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As implantable pumps for continuous delivery of pharmaceutical agents are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg-250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
Specific additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus. For such high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a mid-potency in selected assays described herein (approximately 100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10'8 - 10"4 M of agent should be maintained on the implant or barrier surface. Other specific examples of agents which can be used include those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531
(ruboxistaurin), and simvastatin. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1 % of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10~7 to 10"6 about 10~6 to 10"5 or about 10"5 to 10'4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Although numerous implantable pumps have been described above, all possess similar design features and cause similar unwanted fibrous tissue reactions following implantation and may introduce or promote infection in the area of the implant site. It should be obvious to one of skill in the art that commercial sensor devices not specifically cited above as well as next- generation and/or subsequently-developed commercial implantable pump products are to be anticipated and are suitable for use under the present invention. The clinical function of an implantable drug delivery device or pump depends upon the device, particularly the catheter or drug-dispensing component(s), being able to effectively maintain intimate anatomical contact with the target tissue (e.g., the sudural space in the spinal cord, the arterial lumen, the peritoneum, the interstitial fluid) and not becoming encapsulated or obstructed by scar tissue. For implantable pumps, the drug-delivery catheter lumen, catheter tip, dispensing components, or delivery membrane may become obstructed by scar tissue which may cause the flow of drug to slowdown or cease completely. Alternatively, the entire pump, the catheter and/or the dispensing components can become encapsulated by scar (i.e., the body "walls off" the device with fibrous tissue) so that the drug is incompletely delivered to the target tissue (i.e., the scar prevents proper drug movement and distribution from the implantable pump to the tissues on the other side of the capsule). Either of these developments may lead to inefficient or incomplete drug flow to the desired target tissues or organs (and loss of clinical benefit), while encapsulation can also lead to local drug accumulation (in the capsule) and additional clinical complications (e.g., local drug toxicity; drug sequestration followed by sudden "dumping" of large amounts of drug into the surrounding tissues). For implantable pumps that include electrical or battery components, not only can fibrosis cause the device to function suboptimally or not at all, it can cause excessive drain on battery life as increased energy is required to overcome the increased resistance imposed by the intervening scar tissue. Implantable pumps that release a therapeutic agent for reducing scarring at the device-tissue interface can be used to increase efficacy, prolong clinical performance, ensure that the correct amount of drug is dispensed from the device at the appropriate rate, and reduce the risk that potentially toxic drugs become sequestered in a fibrous capsule. Implantable sensor devices may also benefit from release of a therapeutic agent able to prevent or inhibit infection in the vicinity of the implant site. In one aspect, the present invention provides implantable pumps having the subject compositions infiltrated into adjacent tissue, where the subject compositions may include a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). These compositions may further include one or more fibrosis-inhibiting agents such that the overgrowth of granulation or fibrous tissue is inhibited or reduced and/or one or more anti-infective agents such that infection in the vicinity of the implant site is inhibited or prevented.
4. Sustained-Release Preparations of Fibrosis-inhibiting Agents
As described previously, desired fibrosis-inhibiting agents may be admixed with, blended with, conjugated to, or, otherwise modified to contain a polymer composition (which may be either biodegradable or non- biodegradable), or a non-polymeric composition, in order to release the therapeutic agent over a prolonged period of time. For many of the aforementioned embodiments, localized delivery as well as localized sustained delivery of the fibrosis-inhibiting agent may be required. For example, a desired fibrosis-inhibiting agent may be admixed with, blended with, conjugated to, or otherwise modified to contain a polymeric composition (which may be either biodegradable or non-biodegradable), or non-polymeric composition, in order to release the fibrosis-inhibiting agent over a period of time. In certain aspects, the polymer composition may include a bioerodable or biodegradable polymer. Representative examples of biodegradable polymer compositions suitable for the delivery of fibrosis-inhibiting agents include albumin, collagen, gelatin, hyaluronic acid, starch, cellulose and cellulose derivatives (e.g., methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carboxymethylcellulose, cellulose acetate phthalate, cellulose acetate succinate, hydroxypropylmethylcellulose phthalate), casein, dextrans, polysaccharides, fibrinogen, poly(ether ester) multiblock copolymers, based on poly(ethylene glycol) and polyφutylene terephthalate), tyrosine-derived polycarbonates (e.g., U.S. Patent No. 6,120,491), poly(hydroxyl acids), poly(D,L-lactide), poly(D,L-lactide-co-glycolide), poly(glycolide), poly(hydroxybutyrate), polydioxanone, poly(alkylcarbonate) and poly(orthoesters), polyesters, poly(hydroxy valeric acid), polydioxanone, poly(ethylene terephthalate), poly(malic acid), poly(tartronic acid), poly(acrylamides), polyanhydrides, polyphosphazenes, poly(amino acids), poly(alkylene oxide)-poly(ester) block copolymers (e.g. , X-Y, X-Y-X or Y-X-Y, where X is a polyalkylene oxide and Y is a polyester (e.g., PLGA, PLA, PCL, polydioxanone and copolymers thereof) and their copolymers as well as blends thereof, (see generally, Ilium, L., Davids, S. S. (eds.) "Polymers in Controlled Drug Delivery" Wright, Bristol, 1987; Arshady, J. Controlled Release 77:1-22, 1991 ; Pitt, Int. J. Phar. 59:173-196, 1990; Holland et al., J. Controlled Release 4:155-0180, 1986). Representative examples of non-degradable polymers suitable for the delivery of fibrosis-inhibiting agents include poly(ethylene-co-vinyl acetate) ("EVA") copolymers, silicone rubber, acrylic polymers (polyacrylic acid, polymethylacrylic acid, polymethylmethacrylate, poly(butyl methacrylate)), poly(alkylcynoacrylate) (e.g., poly(ethylcyanoacrylate), poly(butylcyanoacrylate) poly(hexylcyanoacrylate) poly(octylcyanoacrylate)), polyethylene, polypropylene, polyamides (nylon 6,6), polyurethane, poly(ester urethanes), poly(ether urethanes), poly(ester-urea), polyethers (poly(ethylene oxide), poly(propylene oxide), block copolymers based on ethylene oxide and propylene oxide (Le. t copolymers of ethylene oxide and propylene oxide polymers), such as the family of PLURONIC polymers available from BASF Corporation (Mount Olive, NJ), and poly(tetramethylene glycol)), styrene-based polymers (polystyrene, poly(styrene sulfonic acid), poly(styrene)-block- poly(isobutylene)-block-poly(styrene), poly(styrene)-poly(isoprene) block copolymers), and vinyl polymers (polyvinylpyrrolidone, polyvinyl alcohol), polyvinyl acetate phthalate) as well as copolymers and blends thereof. Polymers may also be developed which are either anionic (e.g., alginate, carrageenan, carboxymethyl cellulose, poly(acrylamido-2-methyl propane sulfonic acid) and copolymers thereof, poly(methacrylic acid and copolymers thereof and poly(acrylic acid) and copolymers thereof, as well as blends thereof, or cationic (e.g., chitosan, poly-L-lysine, polyethylenimine, and poly(allyl amine)) and blends thereof (see generally, Dunn et al., J. Applied Polymer ScL 50:353-365, 1993; Cascone et al., J. Materials Sci.: Materials in Medicine 5:770-774, 1994; Shiraishi et al., Biol. Pharm. Bull. 76(11):1164-1168, 1993; Thacharodi and Rao, Int'I J. Pharm. 120A ^S-WQ, 1995; Miyazaki et al., Int'I J. Pharm. 118:257-263, 1995). Particularly preferred polymeric carriers include poly(ethylene-co- vinyl acetate), polyurethanes, poly (D,L-lactic acid) oligomers and polymers, poly (L-lactic acid) oligomers and polymers, poly (glycolic acid), copolymers of lactic acid and glycolic acid, poly (caprolactone), poly (valerolactone), polyanhydrides, copolymers of poly (caprolactone) or poly (lactic acid) with a polyethylene glycol (e.g., MePEG), silicone rubbers, poly(styrene)block- poly(isobutylene)-block-poly(styrene), poly(acrylate) polymers and blends, admixtures, or co-polymers of any of the above. Other preferred polymers include collagen, poly(alkylene oxide)-based polymers, polysaccharides such as hyaluronic acid, chitosan and fucans, and copolymers of polysaccharides with degradable polymers.
Other representative polymers capable of sustained localized delivery of fibrosis-inhibiting agents include carboxylic polymers, polyacetates, polyacrylamides, polycarbonates, polyethers, polyesters, polyethylenes, polyvinylbutyrals, polysilanes, polyureas, polyurethanes, polyoxides, polystyrenes, polysulfides, polysulfones, polysulfonides, polyvinylhalides, pyrrolidones, rubbers, thermal-setting polymers, cross-linkable acrylic and methacrylic polymers, ethylene acrylic acid copolymers, styrene acrylic copolymers, vinyl acetate polymers and copolymers, vinyl acetal polymers and copolymers, epoxy, melamine, other amino resins, phenolic polymers, and copolymers thereof, water-insoluble cellulose ester polymers (including cellulose acetate propionate, cellulose acetate, cellulose acetate butyrate, cellulose nitrate, cellulose acetate phthalate, and mixtures thereof), polyvinylpyrrolidone, polyethylene glycols, polyethylene oxide, polyvinyl alcohol, polyethers, polysaccharides, hydrophilic polyurethane, polyhydroxyacrylate, dextran, xanthan, hydroxypropyl cellulose, methyl cellulose, and homopolymers and copolymers of N-vinylpyrrolidone, N-vinyllactam, N-vinyl butyrolactam, N- vinyl caprolactam, other vinyl compounds having polar pendant groups, acrylate and methacrylate having hydrophilic esterifying groups, hydroxyacryiate, and acrylic acid, and combinations thereof; cellulose esters and ethers, ethyl cellulose, hydroxyethyl cellulose, cellulose nitrate, cellulose acetate, cellulose acetate butyrate, cellulose acetate propionate, polyurethane, polyacrylate, natural and synthetic elastomers, rubber, acetal, nylon, polyester, styrene polybutadiene, acrylic resin, polyvinylidene chloride, polycarbonate, homopolymers and copolymers of vinyl compounds, polyvinylchloride, polyvinylchloride acetate. Representative examples of patents relating to drug-delivery polymers and their preparation include PCT Publication Nos. WO 98/19713, WO 01/17575, WO 01/41821, WO 01/41822, and WO 01/15526 (as well as their corresponding U.S. applications), and U.S. Patent Nos. 4,500,676, 4,582,865, 4,629,623, 4,636,524, 4,713,448, 4,795,741 , 4,913,743, 5,069,899, 5,099,013, 5,128,326, 5,143,724, 5,153,174, 5,246,698, 5,266,563, 5,399,351, 5,525,348, 5,800,412, 5,837,226, 5,942,555, 5,997,517, 6,007,833, 6,071 ,447, 6,090,995, 6,106,473, 6,110,483, 6,121 ,027, 6,156,345, 6,214,901 , 6,368,611 6,630,155, 6,528,080, RE37.950, 6,46,1631, 6,143,314, 5,990,194, 5,792,469, 5,780,044, 5,759,563, 5,744,153, 5,739,176, 5,733,950, 5,681 ,873, 5,599,552, 5,340,849, 5,278,202, 5,278,201 , 6,589,549, 6,287,588, 6,201 ,072, 6,117,949, 6,004,573, 5,702,717, 6,413,539, and 5,714,159, 5,612,052 and U.S. Patent Application Publication Nos. 2003/0068377, 2002/0192286, 2002/0076441, and 2002/0090398.
It may be obvious to one of skill in the art that the polymers as described herein can also be blended or copolymerized in various compositions as required to deliver therapeutic doses of fibrosis-inhibiting agents.
Polymeric carriers for fibrosis-inhibiting agents can be fashioned in a variety of forms, with desired release characteristics and/or with specific properties depending upon the device, composition or implant being utilized. For example, polymeric carriers may be fashioned to release a fibrosis- inhibiting agent upon exposure to a specific triggering event such as pH (see, e.g., Heller et al., "Chemically Self-Regulated Drug Delivery Systems," in Polymers in Medicine III, Elsevier Science Publishers B.V., Amsterdam, 1988, pp. 175-188; Kang et al., J. Applied Polymer Sci. 48:343-354, 1993; Dong et al., J. Controlled Release 19:171-178, 1992; Dong and Hoffman, J. Controlled Release 75:141-152, 1991 ; Kim et al., J. Controlled Release 28:143-152, 1994; Cornejo-Bravo et al., J. Controlled Release 33:223-229, 1995; Wu and Lee, Pharm. Res. -/0(10): 1544-1547, 1993; Serres et al., Pharm. Res. 73(2):196- 201 , 1996; Peppas, "Fundamentals of pH- and Temperature-Sensitive Delivery Systems," in Gurny et al. (eds.), Pulsatile Drug Delivery, Wissenschaftliche Verlagsgesellschaft mbH, Stuttgart, 1993, pp. 41-55; Doelker, "Cellulose Derivatives," 1993, in Peppas and Langer (eds.), Biopolymers I, Springer- Verlag, Berlin). Representative examples of pH-sensitive polymers include poly(acrylic acid) and its derivatives (including for example, homopolymers such as poly(aminocarboxylic acid); poly(acrylic acid); poly(methyl acrylic acid), copolymers of such homopolymers, and copolymers of poly(acrylic acid) and/or acrylate or acrylamide lmonomers such as those discussed above. Other pH sensitive polymers include polysaccharides such as cellulose acetate phthalate; hydroxypropylmethylcellulose phthalate; hydroxypropylmethylcellulose acetate succinate; cellulose acetate trimellilate; and chitosan. Yet other pH sensitive polymers include any mixture of a pH sensitive polymer and a water-soluble polymer. Likewise, fibrosis-inhibiting agents can be delivered via polymeric carriers which are temperature sensitive (see, e.g., Chen et al., "Novel Hydrogels of a Temperature-Sensitive PLURONIC Grafted to a Bioadhesive Polyacrylic Acid Backbone for Vaginal Drug Delivery," in Proceed. Intern. Symp. Control. ReI. Bioact Mater. 22:167-168, Controlled Release Society, Inc., 1995; Okano, "Molecular Design of Stimuli-Responsive Hydrogels for Temporal Controlled Drug Delivery," in Proceed. Intern. Symp. Control. ReI. Bioact. Mater. 22:111-112, Controlled Release Society, Inc., 1995; Johnston et al., Pharm. Res. 9(3):425-433, 1992; Tung, InVI J. Pharm. 707:85-90, 1994; Harsh and Gehrke, J. Controlled Release 77:175-186, 1991; Bae et al., Pharm. Res. 8(4):531-537, 1991; Dinarvand and DΕmanuele, J. Controlled Release 36:221- 227, 1995; Yu and Grainger, "Novel Thermo-sensitive Amphophilic Gels: Poly N- isopropylacrylamide-co-sodium acrylate-co-n-N-alkylacrylamide Network Synthesis and Physicochemical Characterization," Dept. of Chemical & Biological ScL, Oregon Graduate Institute of Science & Technology, Beaverton, OR, pp. 820-821 ; Zhou and Smid, "Physical Hydrogels of Associative Star Polymers," Polymer Research Institute, Dept. of Chemistry, College of Environmental Science and Forestry, State Univ. of New York, Syracuse, NY, pp. 822-823; Hoffman et al., "Characterizing Pore Sizes and Water 'Structure' in Stimuli-Responsive Hydrogels," Center for Bioengineering, Univ. of
Washington, Seattle, WA, p. 828; Yu and Grainger, "Thermo-sensitive Swelling Behavior in Crosslinked N-isopropylacrylamide Networks: Cationic, Anionic and Ampholytic Hydrogels," Dept. of Chemical & Biological ScL, Oregon Graduate Institute of Science & Technology, Beaverton, OR, pp. 829-830; Kim et al., Pharm. Res. 9(3):283-290, 1992; Bae et al., Pharm. Res. 8(5):624-628, 1991 ; Kono et al., J. Controlled Release 30:69-75, 1994; Yoshida et al., J. Controlled Release 32:97-102, 1994; Okano et al., J. Controlled Release 36:125-133, 1995; Chun and Kim, J. Controlled Release 38:39-47, 1996; DΕmanuele and Dinarvand, InVI J. Pharm. 118:237-242, 1995; Katono et al., J. Controlled Release 76:215-228, 1991 ; Hoffman, "Thermally Reversible Hydrogels
Containing Biologically Active Species," in Migliaresi et al. (eds.), Polymers in Medicine III, Eisevier Science Publishers B.V., Amsterdam, 1988, pp. 161-167; Hoffman, "Applications of Thermally Reversible Polymers and Hydrogels in Therapeutics and Diagnostics," in Third International Symposium on Recent Advances in Drug Delivery Systems, Salt Lake City, UT, Feb. 24-27, 1987, pp. 297-305; Gutowska et al., J. Controlled Release 22:95-104, 1992; Palasis and Gehrke, J. Controlled Release 78:1-12, 1992; Paavola et al., Pharm. Res. 12(12):1997-2002, 1995).
Representative examples of thermogelling polymers, and their gelatin temperature (LCST (°C)) include homopolymers such as poly(N-methyl-N-n-propylacrylamide), 19.8; poly(N-n-propylacrylamide), 21.5; poly(N-methyl-N-isopropylacrylamide), 22.3; poly(N-n-propylrnethacrylamide), 28.0; poly(N-isopropylacrylamide), 30.9; poly(N, n-diethylacrylamide), 32.0; poly(N-isopropylmethacrylamide), 44.0; poly^-cyclopropylacrylamide), 45.5; poly(N-ethylmethyacrylamide), 50.0; poly(N-methyl-N-ethylacrylamide), 56.0; poly^-cyclopropylmethacrylamide), 59.0; poly(N-ethylacrylamide), 72.0. Moreover thermogelling polymers may be made by preparing copolymers between (among) monomers of the above, or by combining such homopolymers with other water-soluble polymers such as acrylmonomers (e.g., acrylic acid and derivatives thereof, such as methylacrylic acid, acrylate monomers and derivatives thereof, such as butyl methacrylate, butyl acrylate, lauryl acrylate, and acrylamide monomers and derivatives thereof, such as N-butyl acrylamide and acrylamide).
Other representative examples of thermogelling polymers include cellulose ether derivatives such as hydroxypropyl cellulose, 410C; methyl cellulose, 550C; hydroxypropylmethyl cellulose, 66°C; and ethylhydroxyethyl cellulose, polyalkylene oxide-polyester block copolymers of the structure X-Y, Y-X-Y and X-Y-X where X in a polyalkylene oxide and Y is a biodegradable polyester (e.g., PLG-PEG-PLG) and PLURONICs such as F-127, 10 - 15°C; L-122, 19°C; L-92, 26°C; L-81 , 2O0C; and L-61 , 24°C. Representative examples of patents relating to thermally gelling polymers and their preparation include U.S. Patent Nos. 6,451,346; 6,201,072; 6,117,949; 6,004,573; 5,702,717; and 5,484,610 and PCT Publication Nos. WO 99/07343; WO 99/18142; WO 03/17972; WO 01/82970; WO 00/18821 ; WO 97/15287; WO 01/41735; WO 00/00222 and WO 00/38651.
Fibrosis-inhibiting agents may be linked by occlusion in the matrices of the polymer, bound by covalent linkages, or encapsulated in microcapsules. Within certain embodiments of the invention, therapeutic compositions are provided in non-capsular formulations such as microspheres (ranging from nanometers to micrometers in size), pastes, threads of various size, films and sprays. Within certain aspects of the present invention, therapeutic compositions may be fashioned into particles having any size ranging from 50 nm to 500 μm, depending upon the particular use. These compositions can be in the form of microspheres, microparticles and/or nanoparticles. These compositions can be formed by spray-drying methods, milling methods, coacervation methods, W/O emulsion methods, W/O/W emulsion methods, and solvent evaporation methods. In another embodiment, these compositions can include microemulsions, emulsions, liposomes and micelles. Alternatively, such compositions may also be readily applied as a "spray", which solidifies into a film or coating for use as a device/implant surface coating or to line the tissues of the implantation site. Such sprays may be prepared from microspheres of a wide array of sizes, including for example, from 0.1 μm to 3 μm, from 10 μm to 30 μm, and from 30 μm to 100 μm.
Therapeutic compositions of the present invention may also be prepared in a variety of paste or gel forms. For example, within one embodiment of the invention, therapeutic compositions are provided which are liquid at one temperature (e.g., temperature greater than 37°C, such as 400C, 45°C, 5O0C, 55°C or 6O0C), and solid or semi-solid at another temperature (e.g., ambient body temperature, or any temperature lower than 37°C). Such "thermopastes" may be readily made utilizing a variety of techniques (see, e.g., PCT Publication WO 98/24427). Other pastes may be applied as a liquid, which solidify in vivo due to dissolution of a water-soluble component of the paste and precipitation of encapsulated drug into the aqueous body environment. These "pastes" and "gels" containing fibrosis-inhibiting agents are particularly useful for application to the surface of tissues that will be in contact with the implant or device. Within yet other aspects of the invention, the therapeutic compositions of the present invention may be formed as a film or tube. These films or tubes can be porous or non-porous. Such films or tubes are generally less than 5, 4, 3, 2, or 1 mm thick, or less than 0.75 mm, or less than 0.5 mm, or less than 0.25 mm, or, less than 0.10 mm thick. Films or tubes can also be generated of thicknesses less than 50 μm, 25 μm or 10 μm. Such films may be flexible with a good tensile strength (e.g., greater than 50, or greater than 100, or greater than 150 or 200 N/cm2), good adhesive properties (i.e., adheres to moist or wet surfaces), and have controlled permeability. Fibrosis-inhibiting agents contained in polymeric films are particularly useful for application to the surface of a device or implant as well as to the surface of tissue, cavity or an organ.
Within further aspects of the present invention, polymeric carriers are provided which are adapted to contain and release a hydrophobic fibrosis- inhibiting compound, and/or the carrier containing the hydrophobic compound in combination with a carbohydrate, protein or polypeptide. Within certain embodiments, the polymeric carrier contains or comprises regions, pockets, or granules of one or more hydrophobic compounds. For example, within one embodiment of the invention, hydrophobic compounds may be incorporated within a matrix which contains the hydrophobic fibrosis-inhibiting compound, followed by incorporation of the matrix within the polymeric carrier. A variety of matrices can be utilized in this regard, including for example, carbohydrates and polysaccharides such as starch, cellulose, dextran, methylcellulose, sodium alginate, heparin, chitosan, hyaluronic acid, proteins or polypeptides such as albumin, collagen and gelatin. Within alternative embodiments, hydrophobic compounds may be contained within a hydrophobic core, and this core contained within a hydrophilic shell. Other carriers that may likewise be utilized to contain and deliver fibrosis-inhibiting agents described herein include: hydroxypropyl cyclodextrin (Cserhati and Hollo, Int. J. Pharm. 108:69-75, 1994), liposomes (see, e.g., Sharma et al., Cancer Res. 53:5877-5881, 1993; Sharma and Straubinger, Pharm. Res. 77(60):889-896, 1994; WO 93/18751 ; U.S. Patent No. 5,242,073), liposome/gel (WO 94/26254), nanocapsules (Bartoli et al., J. Microencapsulation 7(2):191-197, 1990), micelles (Alkan-Onyuksel et al., Pharm. Res. 77(2):206-212, 1994), implants (Jampel et al., Invest. Ophthalm. Vis. Science 34(11):3076-3083, 1993; Walter et al., Cancer Res. 54:22017- 2212, 1994), nanoparticles (Violante and Lanzafame PAACR), nanoparticles - modified (U.S. Patent No. 5,145,684), nanoparticles (surface modified) (U.S. Patent No. 5,399,363), micelle (surfactant) (U.S. Patent No. 5,403,858), synthetic phospholipid compounds (U.S. Patent No. 4,534,899), gas borne dispersion (U.S. Patent No. 5,301 ,664), liquid emulsions, foam, spray, gel, lotion, cream, ointment, dispersed vesicles, particles or droplets solid- or liquid- aerosols, microemulsions (U.S. Patent No. 5,330,756), polymeric shell (nano- and micro- capsule) (U.S. Patent No. 5,439,686), emulsion (Tarr et al., Pharm Res. 4: 62-165, 1987), nanospheres (Hagan et al., Proc. Intern. Sy mp. Control ReI. Bioact. Mater. 22, 1995; Kwon et al., Pharm Res. 72(2):192-195; Kwon et al., Pharm Res. 10(7): 970-974; Yokoyama et al., J. Contr. ReI. 32:269-277, 1994; Gref et al., Science 263:1600-1603, 1994; Bazile et al., J. Pharm. Sci. 84:493-498, 1994) and implants (U.S. Patent No. 4,882,168).
Within another aspect of the present invention, polymeric carriers can be materials that are formed in situ. In one embodiment, the precursors can be monomers or macromers that contain unsaturated groups that can be polymerized and/or cross-linked. The monomers or macromers can then, for example, be injected into the treatment area or onto the surface of the treatment area and polymerized in situ using a radiation source (e.g., visible light, UV light) or a free radical system (e.g., potassium persulfate and ascorbic acid or iron and hydrogen peroxide). The polymerization step can be performed immediately prior to, simultaneously to or post injection of the reagents into the treatment site. Representative examples of compositions that undergo free radical polymerization reactions are described in WO 01/44307, WO 01/68720, WO 02/072166, WO 03/043552, WO 93/17669, WO 00/64977, U.S. Patent Nos. 5,900,245, 6,051 ,248, 6,083,524, 6,177,095, 6,201,065, 6,217,894, 6,639,014, 6,352,710, 6,410,645, 6,531 ,147, 5,567,435, 5,986,043, 6,602,975, and U.S. Patent Application Publication Nos. 2002/012796A1 , 2002/0127266A1, 2002/0151650A1 , 2003/0104032A1 , 2002/0091229A1, and 2003/0059906A1.
In another embodiment, the reagents can undergo an electrophilic-nucleophilic reaction to produce a crosslinked matrix. For example, a 4-armed thiol derivatized polyethylene glycol can be reacted with a 4 armed NHS-derivatized polyethylene glycol under basic conditions (pH > about 8). Representative examples of compositions that undergo electrophilic- nucleophilic crosslinking reactions are described in U.S. Patent. Nos. 5,752,974; 5,807,581 ; 5,874,500; 5,936,035; 6,051 ,648; 6,165,489; 6,312,725; 6,458,889; 6,495,127; 6,534,591 ; 6,624,245; 6,566,406; 6,610,033; 6,632,457; U.S. Patent Application Publication No. 2003/0077272; and PCT Application Publication Nos. WO 04/060405 and WO 04/060346. Other examples of in situ forming materials that can be used include those based on the crosslinking of proteins (described in U.S. Patent Nos. RE38158; 4,839,345; 5,514,379, 5,583,114; 6,458,147; 6,371 ,975; U.S. Patent Application Publication Nos 2002/0161399; 2001/0018598 and PCT Publication Nos. WO 03/090683; WO 01/45761 ; WO 99/66964 and WO 96/03159).
The following further and additionally describes polymeric crosslinked matrices, and polymeric carriers, that may be used to assist in the prevention of the formation or growth of fibrous connective tissue. The composition may contain and deliver fibrosis-inhibiting agents in the vicinity of the medical device. The following compositions are particularly useful when it is desired to infiltrate around the device, with or without a fibrosis-inhibiting agent. Such polymeric materials may be prepared from, e.g., (a) synthetic materials, (b) naturally-occurring materials, or (c) mixtures of synthetic and naturally occurring materials. The matrix may be prepared from, e.g., (a) a one- component, i.e., self-reactive, compound, or (b) two or more compounds that are reactive with one another. Typically, these materials are fluid prior to delivery, and thus can be sprayed or otherwise extruded from a device in order to deliver the composition. After delivery, the component materials react with each other, and/or with the body, to provide the desired affect. In some instances, materials that are reactive with one another must be kept separated prior to delivery to the patient, and are mixed together just prior to being delivered to the patient, in order that they maintain a fluid form prior to delivery. In a preferred aspect of the invention, the components of the matrix are delivered in a liquid state to the desired site in the body, whereupon in situ polymerization occurs.
First and Second Synthetic Polymers
In one embodiment, crosslinked polymer compositions (in other words, crosslinked matrices) are prepared by reacting a first synthetic polymer containing two or more nucleophilic groups with a second synthetic polymer containing two or more electrophilic groups, where the electrophilic groups are capable of covalently binding with the nucleophilic groups. In one embodiment, the first and second polymers are each non-immunogenic. In another embodiment, the matrices are not susceptible to enzymatic cleavage by, e.g., a matrix metalloproteinase (e.g., collagenase) and are therefore expected to have greater long-term persistence in vivo than collagen-based compositions.
As used herein, the term "polymer" refers inter alia to polyalkyls, polyamino acids, polyalkyleneoxides and polysaccharides. Additionally, for external or oral use, the polymer may be polyacrylic acid or carbopol. As used herein, the term "synthetic polymer" refers to polymers that are not naturally occurring and that are produced via chemical synthesis. As such, naturally occurring proteins such as collagen and naturally occurring polysaccharides such as hyaluronic acid are specifically excluded. Synthetic collagen, and synthetic hyaluronic acid, and their derivatives, are included. Synthetic polymers containing either nucleophilic or electrophilic groups are also referred to herein as "multifunctionally activated synthetic polymers." The term "multifunctionally activated" (or, simply, "activated") refers to synthetic polymers which have, or have been chemically modified to have, two or more nucleophilic or electrophilic groups which are capable of reacting with one another (i.e., the nucleophilic groups react with the electrophilic groups) to form covalent bonds. Types of multifunctionally activated synthetic polymers include difunctionally activated, tetrafunctionally activated, and star-branched polymers.
Multifunctionally activated synthetic polymers for use in the present invention must contain at least two, more preferably, at least three, functional groups in order to form a three-dimensional crosslinked network with synthetic polymers containing multiple nucleophilic groups (i.e., "multi- nucleophilic polymers"). In other words, they must be at least difunctionally activated, and are more preferably trifunctionally or tetrafunctionally activated. If the first synthetic polymer is a difunctionally activated synthetic polymer, the second synthetic polymer must contain three or more functional groups in order to obtain a three-dimensional crosslinked network. Most preferably, both the first and the second synthetic polymer contain at least three functional groups. Synthetic polymers containing multiple nucleophilic groups are also referred to generically herein as "multi-nucleophilic polymers." For use in the present invention, multi-nucleophilic polymers must contain at least two, more preferably, at least three, nucleophilic groups. If a synthetic polymer containing only two nucleophilic groups is used, a synthetic polymer containing three or more electrophilic groups must be used in order to obtain a three- dimensional crosslinked network.
Preferred multi-nucleophilic polymers for use in the compositions and methods of the present invention include synthetic polymers that contain, or have been modified to contain, multiple nucleophilic groups such as primary amino groups and thiol groups. Preferred multi-nucleophilic polymers include: (i) synthetic polypeptides that have been synthesized to contain two or more primary amino groups or thiol groups; and (ii) polyethylene glycols that have been modified to contain two or more primary amino groups or thiol groups. In general, reaction of a thiol group with an electrophilic group tends to proceed more slowly than reaction of a primary amino group with an electrophilic group.
In one embodiment, the multi-nucleophilic polypeptide is a synthetic polypeptide that has been synthesized to incorporate amino acid residues containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine). Poly(lysine), a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred. Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000.
Poly(lysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000; more preferably, within the range of about 5,000 to about 100,000; most preferably, within the range of about 8,000 to about 15,000. Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.) and Aldrich Chemical (Milwaukee, Wl).
Polyethylene glycol can be chemically modified to contain multiple primary amino or thiol groups according to methods set forth, for example, in Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, N.Y. (1992). Polyethylene glycols which have been modified to contain two or more primary amino groups are referred to herein as "multi-amino PEGs." Polyethylene glycols which have been modified to contain two or more thiol groups are referred to herein as "multi-thiol PEGs." As used herein, the term "polyethylene glycol(s)" includes modified and or derivatized polyethylene glycol(s).
Various forms of multi-amino PEG are commercially available from Shearwater Polymers (Huntsville, Ala.) and from Huntsman Chemical Company (Utah) under the name "Jeffamine." Multi-amino PEGs useful in the present invention include Huntsman's Jeffamine diamines ("D" series) and triamines ("T" series), which contain two and three primary amino groups per molecule, respectively.
Polyamines such as ethylenediamine (H2N-CH2-CH2-NH2), tetramethylenediamine (H2N-(CH2)4-NH2), pentamethylenediamine (cadaverine) (H2N-(CH2)5-NH2), hexamethylenediamine (H2N-(CH2)6-NH2), di(2- aminoethyl)amine (HN-(CH2-CH2-NH2)2), and tris(2-aminoethyl)amine (N-(CH2- CH2-NH2)3) may also be used as the synthetic polymer containing multiple nucleophilic groups.
Synthetic polymers containing multiple electrophilic groups are also referred to herein as "multi-electrophilic polymers." For use in the present invention, the multifunctionally activated synthetic polymers must contain at least two, more preferably, at least three, eiectrophilic groups in order to form a three-dimensional crosslinked network with multi-nucleophilic polymers. Preferred multi-electrophilic polymers for use in the compositions of the invention are polymers which contain two or more succinimidyl groups capable of forming covalent bonds with nucleophilic groups on other molecules. Succinimidyl groups are highly reactive with materials containing primary amino (NH2) groups, such as multi-amino PEG, poly(lysine), or collagen. Succinimidyl groups are slightly less reactive with materials containing thiol (SH) groups, such as multi-thiol PEG or synthetic polypeptides containing multiple cysteine residues.
As used herein, the term "containing two or more succinimidyl groups" is meant to encompass polymers which are preferably commercially available containing two or more succinimidyl groups, as well as those that must be chemically derivatized to contain two or more succinimidyl groups. As used herein, the term "succinimidyl group" is intended to encompass sulfosuccinimidyl groups and other such variations of the "generic" succinimidyl group. The presence of the sodium sulfite moiety on the sulfosuccinimidyl group serves to increase the solubility of the polymer. Hydrophilic polymers and, in particular, various derivatized polyethylene glycols, are preferred for use in the compositions of the present invention. As used herein, the term "PEG" refers to polymers having the repeating structure (OCH2-CHa)n- Structures for some specific, tetrafunctionally activated forms of PEG are shown in FIGS. 4 to 13 of U.S. Patent 5,874,500, incorporated herein by reference. Examples of suitable PEGS include PEG succinimidyl propionate (SE-PEG), PEG succinimidyl succinamide (SSA-PEG), and PEG succinimidyl carbonate (SC-PEG). In one aspect of the invention, the crosslinked matrix is formed in situ by reacting pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG) as reactive reagents. Structures for these reactants are shown in U.S. Patent 5,874,500. Each of these materials has a core with a structure that may be seen by adding ethylene oxide-derived residues to each of the hydroxyl groups in pentaerythritol, and then derivatizing the terminal hydroxyl groups (derived from the ethylene oxide) to contain either thiol groups (so as to form 4-armed thiol PEG) or N-hydroxysuccinimydyl groups (so as to form 4-armed NHS
PEG), optionally with a linker group present between the ethylene oxide derived backbone and the reactive functional group, where this product is commercially available as COSEAL from Angiotech Pharmaceuticals Inc. Optionally, a group "D" may be present in one or both of these molecules, as discussed in more detail below.
As discussed above, preferred activated polyethylene glycol derivatives for use in the invention contain succinimidyl groups as the reactive group. However, different activating groups can be attached at sites along the length of the PEG molecule. For example, PEG can be derivatized to form functionally activated PEG propionaldehyde (A-PEG), or functionally activated PEG glycidyl ether (E-PEG), or functionally activated PEG-isocyanate (I-PEG), or functionally activated PEG-vinylsulfone (V-PEG).
Hydrophobic polymers can also be used to prepare the compositions of the present invention. Hydrophobic polymers for use in the present invention preferably contain, or can be derivatized to contain, two or more electrophilic groups, such as succinimidyl groups, most preferably, two, three, or four electrophilic groups. As used herein, the term "hydrophobic polymer" refers to polymers which contain a relatively small proportion of oxygen or nitrogen atoms.
Hydrophobic polymers which already contain two or more succinimidyl groups include, without limitation, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS3), dithiobis^uccinimidylpropionate) (DSP), bis(2-succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'— dithiobis(su!fosuccinimidylpropionate (DTSPP), and their analogs and derivatives. The above-referenced polymers are commercially available from Pierce (Rockford, III.), under catalog Nos. 21555, 21579, 22585, 21554, and 21577, respectively.
Preferred hydrophobic polymers for use in the invention generally have a carbon chain that is no longer than about 14 carbons. Polymers having carbon chains substantially longer than 14 carbons generally have very poor solubility in aqueous solutions and, as such, have very long reaction times when mixed with aqueous solutions of synthetic polymers containing multiple nucleophilic groups.
Certain polymers, such as polyacids, can be derivatized to contain two or more functional groups, such as succinimidyl groups. Polyacids for use in the present invention include, without limitation, trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)-based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid). Many of these polyacids are commercially available from DuPont Chemical Company (Wilmington, DE). According to a general method, polyacids can be chemically derivatized to contain two or more succinimidyl groups by reaction with an appropriate molar amount of N-hydroxysuccinimide (NHS) in the presence of N^N'-dicyclohexylcarbodiimide (DCC).
Polyalcohols such as trimethylolpropane and di(trimethylol propane) can be converted to carboxylic acid form using various methods, then further derivatized by reaction with NHS in the presence of DCC to produce trifunctionally and tetrafunctionally activated polymers, respectively, as described in U.S. application Ser. No. 08/403,358. Polyacids such as heptanedioic acid (HOOC-(CH2)S-COOH), octanedioic acid (HOOC-(CH2)6- COOH), and hexadecanedioic acid (HOOC-(CH2)14-COOH) are derivatized by the addition of succinimidyl groups to produce difunctionally activated polymers. Polyamines such as ethylenediamine, tetramethylenediamine, pentamethylenediamine (cadaverine), hexamethylenediamine, bis (2- aminoethyl)amine, and tris(2-aminoethyl)amine can be chemically derivatized to polyacids, which can then be derivatized to contain two or more succinimidyl groups by reacting with the appropriate molar amounts of N- hydroxysuccinimide in the presence of DCC, as described in U.S. application Ser. No. 08/403,358. Many of these polyamines are commercially available from DuPont Chemical Company.
In a preferred embodiment, the first synthetic polymer will contain multiple nucleophilic groups (represented below as "X") and it will react with the second synthetic polymer containing multiple electrophilic groups (represented below as "Y"), resulting in a covalently bound polymer network, as follows:
Polymer-Xm + Polymer- Yn → Polymer-Z-Polymer wherein m < 2, n ≤ 2, and m + n ≤ 5; where exemplary X groups include -NH2, -SH, -OH, -PH2, CO-NH-
NH2, etc., where the X groups may be the same or different in polymer-Xm; where exemplary Y groups include -CO2-N(COCH2)2, -CO2H, - CHO, -CHOCH2 (epoxide), -N=C=O, -SO2-CH=CH2, -N(COCH)2 (i.e., a five- membered heterocyclic ring with a double bond present between the two CH groups), -S-S-(C5H4N), etc., where the Y groups may be the same or different in polymer-Yπ; and where Z is the functional group resulting from the union of a nucleophilic group (X) and an electrophilic group (Y).
As noted above, it is also contemplated by the present invention that X and Y may be the same or different, i.e., a synthetic polymer may have two different electrophilic groups, or two different nucleophilic groups, such as with glutathione.
In one embodiment, the backbone of at least one of the synthetic polymers comprises alkylene oxide residues, e.g., residues from ethylene oxide, propylene oxide, and mixtures thereof. The term 'backbone' refers to a significant portion of the polymer.
For example, the synthetic polymer containing alkylene oxide residues may be described by the formula X-polymer-X or Y-polymer-Y, wherein X and Y are as defined above, and the term "polymer" represents - (CH2CH2 O)n- or -(CH(CH3)CH2 O)n-.or -(CH2-CH2-O)n-(CH(CH3)CH2-O)n-. In these cases the synthetic polymer may be difunctional.
The required functional group X or Y is commonly coupled to the polymer backbone by a linking group (represented below as "Q"), many of which are known or possible. There are many ways to prepare the various functionalized polymers, some of which are listed below:
Polymer-QrX + Polymer-Q2-Y → Polymer^ -Z-Q2-Polymer
Exemplary Q groups include -O-(CH2)n-; -S-(CH2)n-; -NH-(CH2)n-; -O2C-NH-(CH2)n-; -O2C-(CH2)n-; -O2C-(CR1H)n-; and -0-R2-CO-NH-, which provide synthetic polymers of the partial structures: polymer-O-(CH2)n-(X or Y); polymer-S-(CH2)n-(X or Y); polymer-NH-(CH2)n-(X or Y); polymer-O2C-NH- (CHa)n-(X or Y); polymer-O2C-(CH2)n-(X or Y); polymer-O2C-(CR1 H)n-(X or Y); and polymer-O-R2-CO-NH-(X or Y), respectively. In these structures, n = 1-10, R1 = H or alkyl (i.e., CH3, C2H5, etc.); R2 = CH2, or CO-NH-CH2CH2; and Q1 and Q2 may be the same or different. For example, when Q2 = OCH2CH2 (there is no Qi in this case); Y
= -CO2-N(COCH2)2; and X = -NH2, -SH, or -OH, the resulting reactions and Z groups may be as follows:
Polymer-NH2 + Polymer-O-CH2-CH2-CO2-N(COCH2)2 → Polymer-NH-CO-CH2-CH2-O-Polymer;
Polymer-SH + Polymer-O-CH2-CH2-CO2-N(COCH2)2 → Polymer-S-COChbCI-b-O-Polymer; and
Polymer-OH + Polymer-O-CH2-CH2-CO2-N(COCH2)2 → Polymer-O-COCH2CH2-O-Polymer.
An additional group, represented below as "D", can be inserted between the polymer and the linking group, if present. One purpose of such a D group is to affect the degradation rate of the crosslinked polymer composition in vivo, for example, to increase the degradation rate, or to decrease the degradation rate. This may be useful in many instances, for example, when drug has been incorporated into the matrix, and it is desired to increase or decrease polymer degradation rate so as to influence a drug delivery profile in the desired direction. An illustration of a crosslinking reaction involving first and second synthetic polymers each having D and Q groups is shown below.
Polymer-D-Q-X + Polymer-D-Q-Y → Polymer-D-Q-Z-Q-D-Polymer
Some useful biodegradable groups "D" include polymers formed from one or more α-hydroxy acids, e.g., lactic acid, glycolic acid, and the cyclization products thereof (e.g., lactide, glycolide), ε-caprolactone, and amino acids. The polymers may be referred to as polylactide, polyglycolide, poly(co- lactide-glycolide); poly-ε-caprolactone, polypeptide (also known as poly amino acid, for example, various di- or tri-peptides) and poly(anhydride)s.
In a general method for preparing the crosslinked polymer compositions used in the context of the present invention, a first synthetic polymer containing multiple nucleophilic groups is mixed with a second synthetic polymer containing multiple electrophilic groups. Formation of a three-dimensional crosslinked network occurs as a result of the reaction between the nucleophilic groups on the first synthetic polymer and the electrophilic groups on the second synthetic polymer. The concentrations of the first synthetic polymer and the second synthetic polymer used to prepare the compositions of the present invention will vary depending upon a number of factors, including the types and molecular weights of the particular synthetic polymers used and the desired end use application. In general, when using multi-amino PEG as the first synthetic polymer, it is preferably used at a concentration in the range of about 0.5 to about 20 percent by weight of the final composition, while the second synthetic polymer is used at a concentration in the range of about 0.5 to about 20 percent by weight of the final composition. For example, a final composition having a total weight of 1 gram (1000 milligrams) may contain between about 5 to about 200 milligrams of multi-amino PEG, and between about 5 to about 200 milligrams of the second synthetic polymer. Use of higher concentrations of both first and second synthetic polymers will result in the formation of a more tightly crosslinked network, producing a stiffer, more robust gel. Compositions intended for use in tissue augmentation will generally employ concentrations of first and second synthetic polymer that fall toward the higher end of the preferred concentration range. Compositions intended for use as bioadhesives or in adhesion prevention do not need to be as firm and may therefore contain lower polymer concentrations.
Because polymers containing multiple electrophilic groups will also react with water, the second synthetic polymer is generally stored and used in sterile, dry form to prevent the loss of crosslinking ability due to hydrolysis which typically occurs upon exposure of such electrophilic groups to aqueous media. Processes for preparing synthetic hydrophilic polymers containing multiple electrophilic groups in sterile, dry form are set forth in U.S. Patent 5,643,464. For example, the dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or, preferably, e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates. In contrast, polymers containing multiple nucleophilic groups are generally not water-reactive and can therefore be stored in aqueous solution.
In certain embodiments, one or both of the electrophilic- or nucleophilic-terminated polymers described above can be combined with a synthetic or naturally occurring polymer. The presence of the synthetic or naturally occurring polymer may enhance the mechanical and/or adhesive properties of the in situ forming compositions. Naturally occurring polymers, and polymers derived from naturally occurring polymer that may be included in in situ forming materials include naturally occurring proteins, such as collagen, collagen derivatives (such as methylated collagen), fibrinogen, thrombin, albumin, fibrin, and derivatives of and naturally occurring polysaccharides, such as glycosaminoglycans, including deacetylated and desulfated glycosaminoglycan derivatives.
In one aspect, a composition comprising naturally-occurring protein and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising collagen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising methylated collagen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrinogen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising thrombin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising albumin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising naturally occurring polysaccharide and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising deacetylated glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising desulfated glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
In one aspect, a composition comprising naturally-occurring protein and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising collagen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising methylated collagen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrinogen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising thrombin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising albumin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising naturally occurring polysaccharide and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising deacetylated glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising desulfated glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
In one aspect, a composition comprising naturally-occurring protein and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising collagen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising methylated collagen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrinogen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising thrombin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising albumin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising naturally occurring polysaccharide and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising deacetylated glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising desulfated glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. The presence of protein or polysaccharide components which contain functional groups that can react with the functional groups on multiple activated synthetic polymers can result in formation of a crosslinked synthetic polymer-naturally occurring polymer matrix upon mixing and/or crosslinking of the synthetic polymer(s). In particular, when the naturally occurring polymer (protein or polysaccharide) also contains nucleophilic groups such as primary amino groups, the electrophilic groups on the second synthetic polymer will react with the primary amino groups on these components, as well as the nucleophilic groups on the first synthetic polymer, to cause these other components to become part of the polymer matrix. For example, lysine-rich proteins such as collagen may be especially reactive with electrophilic groups on synthetic polymers.
In one aspect, the naturally occurring protein is polymer may be collagen. As used herein, the term "collagen" or "collagen material" refers to all forms of collagen, including those which have been processed or otherwise modified and is intended to encompass collagen of any type, from any source, including, but not limited to, collagen extracted from tissue or produced recombinantly, collagen analogues, collagen derivatives, modified collagens, and denatured collagens, such as gelatin. In general, collagen from any source may be included in the compositions of the invention; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced. The preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. U.S. Patent No. 5,428,022 discloses methods of extracting and purifying collagen from the human placenta. U.S. Patent No. 5,667,839, discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows. Collagen of any type, including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compositions of the invention, although type I is generally preferred. Either atelopeptide or telopeptide-containing collagen may be used; however, when collagen from a xenogeneic source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen.
Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinkϊng agents is preferred for use in the compositions of the invention, although previously crosslinked collagen may be used. Non-crosslinked atelopeptide fibrillar collagen is commercially available from lnamed Aesthetics (Santa Barbara, CA) at collagen concentrations of 35 mg/ml and 65 mg/ml under the trademarks ZYDERM I Collagen and ZYDERM Il Collagen, respectively. Glutaraldehyde crosslinked atelopeptide fibrillar collagen is commercially available from lnamed Corporation (Santa Barbara, CA) at a collagen concentration of 35 mg/ml under the trademark ZYPLAST Collagen.
Collagens for use in the present invention are generally in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml; preferably, between about 30 mg/ml to about 90 mg/ml.
Because of its tacky consistency, nonfibrillar collagen may be preferred for use in compositions that are intended for use as bioadhesives. The term "nonfibrillar collagen" refers to any modified or unmodified collagen material that is in substantially nonfibrillar form at pH 7, as indicated by optical clarity of an aqueous suspension of the collagen.
Collagen that is already in nonfibrillar form may be used in the compositions of the invention. As used herein, the term "nonfibrillar collagen" is intended to encompass collagen types that are nonfibrillar in native form, as well as collagens that have been chemically modified such that they are in nonfibrillar form at or around neutral pH. Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen and methylated collagen, both of which can be prepared according to the methods described in U.S. Pat. No. 4,164,559, issued Aug. 14, 1979, to Miyata et al., which is hereby incorporated by reference in its entirety. Due to its inherent tackiness, methylated collagen is particularly preferred for use in bioadhesive compositions, as disclosed in U.S. application Ser. No. 08/476,825.
Collagens for use in the crosslinked polymer compositions of the present invention may start out in fibrillar form, then be rendered nonfibrillar by the addition of one or more fiber disassembly agent. The fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above. Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids (e.g., arginine), inorganic salts (e.g., sodium chloride and potassium chloride), and carbohydrates (e.g., various sugars including sucrose).
In one aspect, the polymer may be collagen or a collagen derivative, for example methylated collagen. An example of an in situ forming composition uses pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4- armed thiol PEG), pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG) and methylated collagen as the reactive reagents. This composition, when mixed with the appropriate buffers can produce a crosslinked hydrogel. (See, e.g., U.S. Patent Nos. 5,874,500; 6,051 ,648; 6,166,130; 5,565,519 and 6,312,725;.
In another aspect, the naturally occurring polymer may be a glycosaminoglycan. Giycosaminoglycans, e.g., hyaluronic acid, contain both anionic and cationic functional groups along each polymeric chain, which can form intramolecular and/or intermolecular ionic crosslinks, and are responsible for the thixotropic (or shear thinning) nature of hyaluronic acid.
In certain aspects, the glycosaminoglycan may be derivatized. For example, giycosaminoglycans can be chemically derivatized by, e.g., deacetylation, desulfation, or both in order to contain primary amino groups available for reaction with electrophilic groups on synthetic polymer molecules. Giycosaminoglycans that can be derivatized according to either or both of the aforementioned methods include the following: hyaluronic acid, chondroitin sulfate A, chondroitin sulfate B (dermatan sulfate), chondroitin sulfate C, chitin (can be derivatized to chitosan), keratan sulfate, keratosulfate, and heparin. Derivatization of glycosaminoglycans by deacetylation and/or desulfation and covalent binding of the resulting glycosaminoglycan derivatives with synthetic hydrophilic polymers is described in further detail in commonly assigned, allowed U.S. patent application Ser. No. 08/146,843, filed Nov. 3, 1993.
In general, the collagen is added to the first synthetic polymer, then the collagen and first synthetic polymer are mixed thoroughly to achieve a homogeneous composition. The second synthetic polymer is then added and mixed into the collagen/first synthetic polymer mixture, where it will covalently bind to primary amino groups or thiol groups on the first synthetic polymer and primary amino groups on the collagen, resulting in the formation of a homogeneous crosslinked network. Various deacetylated and/or desulfated glycosaminoglycan derivatives can be incorporated into the composition in a similar manner as that described above for collagen. In addition, the introduction of hydrocolloids such as carboxymethylcellulose may promote tissue adhesion and/or swellability.
Administration of the Crosslinked Synthetic Polymer Compositions
The compositions of the present invention having two synthetic polymers may be administered before, during or after crosslinking of the first and second synthetic polymer. Certain uses, which are discussed in greater detail below, such as tissue augmentation, may require the compositions to be crosslinked before administration, whereas other applications, such as tissue adhesion, require the compositions to be administered before crosslinking has reached "equilibrium." The point at which crosslinking has reached equilibrium is defined herein as the point at which the composition no longer feels tacky or sticky to the touch.
In order to administer the composition prior to crosslinking, the first synthetic polymer and second synthetic polymer may be contained within separate barrels of a dual-compartment syringe. In this case, the two synthetic polymers do not actually mix until the point at which the two polymers are extruded from the tip of the syringe needle into the patient's tissue. This allows the vast majority of the crosslinking reaction to occur in situ, avoiding the problem of needle blockage which commonly occurs if the two synthetic polymers are mixed too early and crosslinking between the two components is already too advanced prior to delivery from the syringe needle. The use of a dual-compartment syringe, as described above, allows for the use of smaller diameter needles, which is advantageous when performing procedures in delicate tissue, such as that surrounding the eyes. Alternatively, the first synthetic polymer and second synthetic polymer may be mixed according to the methods described above prior to delivery to the tissue site, then injected to the desired tissue site immediately (preferably, within about 60 seconds) following mixing.
In another embodiment of the invention, the first synthetic polymer and second synthetic polymer are mixed, then extruded and allowed to crosslink into a sheet or other solid form. The crosslinked solid is then dehydrated to remove substantially all unbound water. The resulting dried solid may be ground or comminuted into particulates, then suspended in a nonaqueous fluid carrier, including, without limitation, hyaluronic acid, dextran sulfate, dextran, succinylated noncrosslinked collagen, methylated noncrosslinked collagen, glycogen, glycerol, dextrose, maltose, triglycerides of fatty acids (such as com oil, soybean oil, and sesame oil), and egg yolk phospholipid. The suspension of particulates can be injected through a small- gauge needle to a tissue site. Once inside the tissue, the crosslinked polymer particulates will rehydrate and swell in size at least five-fold.
Hvdrophilic Polymer + Plurality of Crosslinkable Components
As mentioned above, the first and/or second synthetic polymers may be combined with a hydrophilic polymer, e.g., collagen or methylated collagen, to form a composition useful in the present invention. In one general embodiment, the compositions useful in the present invention include a hydrophilic polymer in combination with two or more crosslinkable components. This embodiment is described in further detail in this section.
The Hvdrophilic Polymer Component:
The hydrophilic polymer component may be a synthetic or naturally occurring hydrophilic polymer. Naturally occurring hydrophilic polymers include, but are not limited to: proteins such as collagen and derivatives therof, fibronectin, albumins, globulins, fibrinogen, and fibrin, with collagen particularly preferred; carboxylated polysaccharides such as polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives. Collagen (e.g., methylated collagen) and glycosaminoglycans are preferred naturally occurring hydrophilic polymers for use herein. In general, collagen from any source may be used in the composition of the method; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced. The preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. See, e.g., U.S. Pat. No. 5,428,022, to Palefsky et al., which discloses methods of extracting and purifying collagen from the human placenta. See also U.S. Patent No. 5,667,839, to Berg, which discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows. Unless otherwise specified, the term "collagen" or "collagen material" as used herein refers to all forms of collagen, including those that have been processed or otherwise modified.
Collagen of any type, including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compositions of the invention, although type I is generally preferred. Either atelopeptide or telopeptide- containing collagen may be used; however, when collagen from a source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen.
Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the compositions of the invention, although previously crosslinked collagen may be used. Non-crosslinked atelopeptide fibrillar collagen is commercially available from McGhan Medical Corporation (Santa Barbara, Calif.) at collagen concentrations of 35 mg/ml and 65 mg/ml under the trademarks ZYDERM® I Collagen and ZYDERM® Il Collagen, respectively. Glutaraldehyde-crosslinked atelopeptide fibrillar collagen is commercially available from McGhan Medical Corporation at a collagen concentration of 35 mg/ml under the trademark ZYPLAST®.
Collagens for use in the present invention are generally, although not necessarily, in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml, preferably between about 30 mg/ml to about 90 mg/ml.
Although intact collagen is preferred, denatured collagen, commonly known as gelatin, can also be used in the compositions of the invention. Gelatin may have the added benefit of being degradable faster than collagen.
Because of its greater surface area and greater concentration of reactive groups, nonfibrillar collagen is generally preferred. The term "nonfibrillar collagen" refers to any modified or unmodified collagen material that is in substantially nonfibrillar form at pH 7, as indicated by optical clarity of an aqueous suspension of the collagen.
Collagen that is already in nonfibrillar form may be used in the compositions of the invention. As used herein, the term "nonfibrillar collagen" is intended to encompass collagen types that are nonfibrillar in native form, as well as collagens that have been chemically modified such that they are in nonfibrillar form at or around neutral pH. Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen, propylated collagen, ethylated collagen, methylated collagen, and the like, both of which can be prepared according to the methods described in U.S. Pat. No. 4,164,559, to Miyata et al., which is hereby incorporated by reference in its entirety. Due to its inherent tackiness, methylated collagen is particularly preferred, as disclosed in U.S. Patent No. 5,614,587 to Rhee et al. Collagens for use in the crosslinkable compositions of the present invention may start out in fibrillar form, then be rendered nonfibrillar by the addition of one or more fiber disassembly agents. The fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above. Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids, inorganic salts, and carbohydrates, with biocompatible alcohols being particularly preferred. Preferred biocompatible alcohols include glycerol and propylene glycol. Non-biocompatible alcohols, such as ethanol, methanol, and isopropanol, are not preferred for use in the present invention, due to their potentially deleterious effects on the body of the patient receiving them. Preferred amino acids include arginine. Preferred inorganic salts include sodium chloride and potassium chloride. Although carbohydrates, such as various sugars including sucrose, may be used in the practice of the present invention, they are not as preferred as other types of fiber disassembly agents because they can have cytotoxic effects in vivo.
As fibrillar collagen has less surface area and a lower concentration of reactive groups than nonfibrillar, fibrillar collagen is less preferred. However, as disclosed in U.S. Patent 5,614,587, fibrillar collagen, or mixtures of nonfibrillar and fibrillar collagen, may be preferred for use in compositions intended for long-term persistence in vivo, if optical clarity is not a requirement. Synthetic hydrophilic polymers may also be used in the present invention. Useful synthetic hydrophilic polymers include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol and poly(ethylene oxide)- poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (particularly highly branched polyglycerol), propylene glycol and trimethylene glycol substituted with one or more polyalkylene oxides, e.g., mono-, di- and tri-polyoxyethylated glycerol, mono- and di-polyoxyethylated propylene glycol, and mono- and di- polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; acrylic acid polymers and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly(hydroxyethyl-methacrylate), poly(hydroxyethylacrylate), poly(methylalkylsulfoxide methacrylate), poly(methylalkylsulfoxide acrylate) and copolymers of any of the foregoing, and/or with additional acrylate species such as aminoethyl acrylate and mono-2-(acryloxy)-ethyl succinate; polymaleic acid; poly(acrylamides) such as polyacrylamide perse, poly(methacrylamide), poly(dimethylacrylamide), and poly(N-isopropyl-acrylamide); poly(olefinic alcohol)s such as polyvinyl alcohol); poly(N-vinyl lactams) such as polyvinyl pyrrolidone), poly(N-vinyl caprolactam), and copolymers thereof; polyoxazolines, including poly(methyloxazoline) and poly(ethyloxazoline); and polyvinylamines. It must be emphasized that the aforementioned list of polymers is not exhaustive, and a variety of other synthetic hydrophilic polymers may be used, as will be appreciated by those skilled in the art.
The Crosslinkable Components: The compositions of the invention also comprise a plurality of crosslinkable components. Each of the crosslinkable components participates in a reaction that results in a crosslinked matrix. Prior to completion of the crosslinking reaction, the crosslinkable components provide the necessary adhesive qualities that enable the methods of the invention. The crosslinkable components are selected so that crosslinking gives rise to a biocompatible, nonimmunogenic matrix useful in a variety of contexts including adhesion prevention, biologically active agent delivery, tissue augmentation, and other applications. The crosslinkable components of the invention comprise: a component A, which has m nucleophilic groups, wherein m > 2 and a component B, which has n electrophilic groups capable of reaction with the m nucleophilic groups, wherein n > 2 and m + n > 4. An optional third component, optional component C, which has at least one functional group that is either electrophilic and capable of reaction with the nucleophilic groups of component A, or nucleophilic and capable of reaction with the electrophilic groups of component B may also be present. Thus, the total number of functional groups present on components A, B and C, when present, in combination is > 5; that is, the total functional groups given by m + n + p must be > 5, where p is the number of functional groups on component C and, as indicated, is > 1. Each of the components is biocompatible and nonimmunogenic, and at least one component is comprised of a hydrophilic polymer. Also, as will be appreciated, the composition may contain additional crosslinkable components D, E, F, etc., having one or more reactive nucleophilic or electrophilic groups and thereby participate in formation of the crosslinked biomaterial via covalent bonding to other components.
The m nucleophilic groups on component A may all be the same, or, alternatively, A may contain two or more different nucleophilic groups. Similarly, the n electrophilic groups on component B may all be the same, or two or more different electrophilic groups may be present. The functional group(s) on optional component C, if nucleophilic, may or may not be the same as the nucleophilic groups on component A, and, conversely, if electrophilic, the functional group(s) on optional component C may or may not be the same as the electrophilic groups on component B.
Accordingly, the components may be represented by the structural formulae
(I) R1(-[Q1]q-X)m (component A), (II) R2(-[Q2]rY)n (component B), and
(III) R3(-[Q3]S-Fn)p (optional component C), wherein:
R1, R2 and R3 are independently selected from the group consisting of C2 to Cu hydrocarbyl, heteroatom-containing C2 to Ci4 hydrocarbyl, hydrophilic polymers, and hydrophobic polymers, providing that at least one of R1, R2 and R3 is a hydrophilic polymer, preferably a synthetic hydrophilic polymer;
X represents one of the m nucleophilic groups of component A, and the various X moieties on A may be the same or different;
Y represents one of the n electrophilic groups of component B, and the various Y moieties on A may be the same or different;
Fn represents a functional group on optional component C; Q1, Q2 and Q3 are linking groups; m > 2, n > 2, m + n is > 4, q, and r are independently zero or 1 , and when optional component C is present, p > 1 , and s is independently zero or 1.
Reactive Groups:
X may be virtually any nucleophilic group, so long as reaction can occur with the electrophilic group Y. Analogously, Y may be virtually any electrophilic group, so long as reaction can take place with X. The only limitation is a practical one, in that reaction between X and Y should be fairly rapid and take place automatically upon admixture with an aqueous medium, without need for heat or potentially toxic or non-biodegradable reaction catalysts or other chemical reagents. It is also preferred although not essential that reaction occur without need for ultraviolet or other radiation. Ideally, the reactions between X and Y should be complete in under 60 minutes, preferably under 30 minutes. Most preferably, the reaction occurs in about 5 to 15 minutes or less. Examples of nucleophilic groups suitable as X include, but are not limited to, -NH2, -NHR4, -N(R4)2, -SH, -OH1 -COOH, -C6H4-OH, -PH2, -PHR5, - P(R5)2, -NH-NH2, -CO-NH-NH2, -C5H4N, etc. wherein R4 and R5 are hydrocarbyl, typically alkyl or monocyclic aryl, preferably alkyl, and most preferably lower alkyl. Organometallic moieties are also useful nucleophilic groups for the purposes of the invention, particularly those that act as carbanion donors. Organometallic nucleophiles are not, however, preferred. Examples of organometallic moieties include: Grignard functionalities -R6MgHaI wherein R6 is a carbon atom (substituted or unsubstituted), and Hal is halo, typically bromo, iodo or chloro, preferably bromo; and lithium-containing functionalities, typically alkyllithium groups; sodium-containing functionalities.
It will be appreciated by those of ordinary skill in the art that certain nucleophilic groups must be activated with a base so as to be capable of reaction with an electrophile. For example, when there are nucleophilic sulfhydryl and hydroxyl groups in the crosslinkable composition, the composition must be admixed with an aqueous base in order to remove a proton and provide an -S" or -O" species to enable reaction with an electrophile. Unless it is desirable for the base to participate in the crosslinking reaction, a nonnucleophilic base is preferred. In some embodiments, the base may be present as a component of a buffer solution. Suitable bases and corresponding crosslinking reactions are described infra.
The selection of electrophilic groups provided within the crosslinkable composition, i.e., on component B, must be made so that reaction is possible with the specific nucleophilic groups. Thus, when the X moieties are amino groups, the Y groups are selected so as to react with amino groups. Analogously, when the X moieties are sulfhydryl moieties, the corresponding electrophilic groups are sulfhydryl-reactive groups, and the like.
By way of example, when X is amino (generally although not necessarily primary amino), the electrophilic groups present on Y are amino reactive groups such as, but not limited to: (1) carboxylic acid esters, including cyclic esters and "activated" esters; (2) acid chloride groups (-CO-CI); (3) anhydrides (-(CO)-O-(CO)-R); (4) ketones and aldehydes, including α,β- unsaturated aldehydes and ketones such as -CH=CH-CH=O and -CH=CH- C(CH3)=O; (5) halides; (6) isocyanate (-N=C=O); (7) isothiocyanate (-N=C=S); (8) epoxides; (9) activated hydroxyl groups (e.g., activated with conventional activating agents such as carbonyldiimidazole or sulfonyl chloride); and (10) olefins, including conjugated olefins, such as ethenesulfonyl (-SO2CH=CH2) and analogous functional groups, including acrylate (-CO2-C=CH2), methacrylate (-CO2-C(CHs)=CH2)), ethyl acrylate (-CO2-C(CH2CHs)=CH2), and ethyleneimino (-CH=CH-C=NH). Since a carboxylic acid group perse is not susceptible to reaction with a nucleophilic amine, components containing carboxylic acid groups must be activated so as to be amine-reactive. Activation may be accomplished in a variety of ways, but often involves reaction with a suitable hydroxyl-containing compound in the presence of a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU). For example, a carboxylic acid can be reacted with an alkoxy-substituted N- hydroxy-succinimide or N-hydroxysulfosuccinimide in the presence of DCC to form reactive electrophilic groups, the N-hydroxysuccinimide ester and the N- hydroxysulfosuccinimide ester, respectively. Carboxylic acids may also be activated by reaction with an acyl halide such as an acyl chloride (e.g., acetyl chloride), to provide a reactive anhydride group. In a further example, a carboxylic acid may be converted to an acid chloride group using, e.g., thionyl chloride or an acyl chloride capable of an exchange reaction. Specific reagents and procedures used to carry out such activation reactions will be known to those of ordinary skill in the art and are described in the pertinent texts and literature.
Analogously, when X is sulfhydryl, the electrophilic groups present on Y are groups that react with a sulfhydryl moiety. Such reactive groups include those that form thioester linkages upon reaction with a sulfhydryl group, such as those described in PCT Publication No. WO 00/62827 to Wallace et al. As explained in detail therein, such "sulfhydryl reactive" groups include, but are not limited to: mixed anhydrides; ester derivatives of phosphorus; ester derivatives of p-nitrophenol, p-nitrothiophenol and pentafluorophenol; esters of substituted hydroxylamines, including N-hydroxyphthalimide esters, N- hydroxysuccinimide esters, N-hydroxysulfosuccinimide esters, and N- hydroxyglutarimide esters; esters of 1-hydroxybenzotriazole; 3-hydroxy-3,4- dihydro-benzotriazin-4-one; 3-hydroxy-3,4-dihydro-quinazoline-4-one; carbonylimidazole derivatives; acid chlorides; ketenes; and isocyanates. With these sulfhydryl reactive groups, auxiliary reagents can also be used to facilitate bond formation, e.g., 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide can be used to facilitate coupling of sulfhydryl groups to carboxyl-containing groups.
In addition to the sulfhydryl reactive groups that form thioester linkages, various other sulfhydryl reactive functionalities can be utilized that form other types of linkages. For example, compounds that contain methyl imidate derivatives form imido-thioester linkages with sulfhydryl groups. Alternatively, sulfhydryl reactive groups can be employed that form disulfide bonds with sulfhydryl groups; such groups generally have the structure -S-S-Ar where Ar is a substituted or unsubstituted nitrogen-containing heteroaromatic moiety or a non-heterocyclic aromatic group substituted with an electron- withdrawing moiety, such that Ar may be, for example, 4-pyridinyl, o- nitrophenyl, m-nitrophenyl, p-nitrophenyl, 2,4-dinitrophenyl, 2-nitro-4-benzoic acid, 2-nitro-4-pyridinyl, etc. In such instances, auxiliary reagents, i.e., mild oxidizing agents such as hydrogen peroxide, can be used to facilitate disulfide bond formation.
Yet another class of sulfhydryl reactive groups forms thioether bonds with sulfhydryl groups. Such groups include, inter alia, maleimido, substituted maleimido, haloalkyl, epoxy, imino, and aziridino, as well as olefins (including conjugated olefins) such as ethenesulfonyl, etheneimino, acrylate, methacrylate, and α,β-unsaturated aldehydes and ketones. This class of sulfhydryl reactive groups is particularly preferred as the thioether bonds may provide faster crosslinking and longer in vivo stability. When X is -OH, the electrophilic functional groups on the remaining component(s) must react with hydroxyl groups. The hydroxyl group may be activated as described above with respect to carboxylic acid groups, or it may react directly in the presence of base with a sufficiently reactive electrophile such as an epoxide group, an aziridine group, an acyl halide, or an anhydride.
When X is an organometallic nucleophile such as a Grignard functionality or an alkyllithium group, suitable electrophilic functional groups for reaction therewith are those containing carbonyl groups, including, by way of example, ketones and aldehydes.
It will also be appreciated that certain functional groups can react as nucleophiles or as electrophiles, depending on the selected reaction partner and/or the reaction conditions. For example, a carboxylic acid group can act as a nucleophile in the presence of a fairly strong base, but generally acts as an electrophile allowing nucleophilic attack at the carbonyl carbon and concomitant replacement of the hydroxyl group with the incoming nucleophile.
The covalent linkages in the crosslinked structure that result upon covalent binding of specific nucleophilic components to specific electrophilic components in the crosslinkable composition include, solely by way of example, the following (the optional linking groups Q1 and Q2 are omitted for clarity):
Table
Figure imgf000363_0001
Figure imgf000364_0001
Figure imgf000365_0001
Linking Groups:
The functional groups X and Y and FN on optional component C may be directly attached to the compound core (R1, R2 or R3 on optional component C, respectively), or they may be indirectly attached through a linking group, with longer linking groups also termed "chain extenders." In structural formulae (I), (II) and (III), the optional linking groups are represented by Q1, Q2 and Q3, wherein the linking groups are present when q, r and s are equal to 1 (with R, X, Y, Fn, m n and p as defined previously).
Suitable linking groups are well known in the art. See, for example, International Patent Publication No. WO 97/22371. Linking groups are useful to avoid steric hindrance problems that are sometimes associated with the formation of direct linkages between molecules. Linking groups may additionally be used to link several multifunctionally activated compounds together to make larger molecules. In a preferred embodiment, a linking group can be used to alter the degradative properties of the compositions after administration and resultant gel formation. For example, linking groups can be incorporated into components A, B, or optional component C to promote hydrolysis, to discourage hydrolysis, or to provide a site for enzymatic degradation. Examples of linking groups that provide hydrolyzable sites, include, inter alia: ester linkages; anhydride linkages, such as obtained by incorporation of glutarate and succinate; ortho ester linkages; ortho carbonate linkages such as trimethylene carbonate; amide linkages; phosphoester linkages; α-hydroxy acid linkages, such as may be obtained by incorporation of lactic acid and glycolic acid; lactone-based linkages, such as may be obtained by incorporation of caprolactone, valerolactone, γ-butyrolactone and p- dioxanone; and amide linkages such as in a dimeric, oligomeric, or poly(amino acid) segment. Examples of non-degradable linking groups include succinimide, propionic acid and carboxymethylate linkages. See, for example, PCT WO 99/07417. Examples of enzymatically degradable linkages include Leu-Gly-Pro-Ala, which is degraded by collagenase; and Gly-Pro-Lys, which is degraded by plasmin.
Linking groups can also enhance or suppress the reactivity of the various nucleophilic and electrophilic groups. For example, electron- withdrawing groups within one or two carbons of a sulfhydryl group may be expected to diminish its effectiveness in coupling, due to a lowering of nucleophilicity. Carbon-carbon double bonds and carbonyl groups will also have such an effect. Conversely, electron-withdrawing groups adjacent to a carbonyl group (e.g., the reactive carbonyl of glutaryl-N-hydroxysuccinimidyl) may increase the reactivity of the carbonyl carbon with respect to an incoming nucleophile. By contrast, sterically bulky groups in the vicinity of a functional group can be used to diminish reactivity and thus coupling rate as a result of steric hindrance. By way of example, particular linking groups and corresponding component structure are indicated in the following Table: Table
Figure imgf000367_0001
In the above Table, n is generally in the range of 1 to about 10, R7 is generally hydrocarbyl, typically alkyl or aryl, preferably alkyl, and most preferably lower alkyl, and R8 is hydrocarbylene, heteroatom-containing hydrocarbylene, substituted hydrocarbylene, or substituted heteroatom- containing hydrocarbylene) typically alkylene or arylene (again, optionally substituted and/or containing a heteroatom), preferably lower alkylene (e.g., methylene, ethylene, n-propylene, n-butylene, etc.), phenylene, or amidoalkylene (e.g., -(CO)-NH-CI-I2).
Other general principles that should be considered with respect to linking groups are as follows: If higher molecular weight components are to be used, they preferably have biodegradable linkages as described above, so that fragments larger than 20,000 mol. wt. are not generated during resorption in the body. In addition, to promote water miscibility and/or solubility, it may be desired to add sufficient electric charge or hydrophilicity. Hydrophilic groups can be easily introduced using known chemical synthesis, so long as they do not give rise to unwanted swelling or an undesirable decrease in compressive strength. In particular, polyalkoxy segments may weaken gel strength.
The Component Core:
The "core" of each crosslinkable component is comprised of the molecular structure to which the nucleophilic or electrophilic groups are bound. Using the formulae (I) R1-[Q1]q-X)m, for component A, (II) R2(-[Q2]rY)n for component B, and (III) R3(-[Q3]S-Fn)p for optional component C, the "core" groups are R1, R2 and R3. Each molecular core of the reactive components of the crosslinkable composition is generally selected from synthetic and naturally occurring hydrophilic polymers, hydrophobic polymers, and C2-Ci4 hydrocarbyl groups zero to 2 heteroatoms selected from N, O and S, with the proviso that at least one of the crosslinkable components A, B, and optionally C, comprises a molecular core of a synthetic hydrophilic polymer. In a preferred embodiment, at least one of A and B comprises a molecular core of a synthetic hydrophilic polymer. Hvdrophilic Crosslinkable Components
In one aspect, the crosslinkable component(s) is (are) hydrophilic polymers. The term "hydrophilic polymer" as used herein refers to a synthetic polymer having an average molecular weight and composition effective to render the polymer "hydrophilic" as defined above. As discussed above, synthetic crosslinkable hydrophilic polymers useful herein include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol and poly(ethylene oxide)-poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (particularly highly branched polyglycerol), propylene glycol and trimethylene glycol substituted with one or more polyalkylene oxides, e.g., mono-, di- and tri-polyoxyethylated glycerol, mono- and di-polyoxyethylated propylene glycol, and mono- and di- polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; acrylic acid polymers and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly(hydroxyethyl-methacrylate), poly(hydroxyethylacrylate), poly(methylalkylsulfoxide methacrylate), poly(methylalkylsulfoxide acrylate) and copolymers of any of the foregoing, and/or with additional acrylate species such as aminoethyl acrylate and mono-2-(acryloxy)-ethyl succinate; polymaleic acid; poly(acrylamides) such as polyacrylamide perse, poly(methacrylamide), poly(dimethylacrylamide), and poly(N-isopropyl-acrylamide); poly(olefinic alcohol)s such as polyvinyl alcohol); poly(N-vinyl lactams) such as polyvinyl pyrrolidone), poly(N-vinyl caprolactam), and copolymers thereof; polyoxazolines, including poly(methyloxazoline) and poly(ethyloxazoline); and polyvinylamines. It must be emphasized that the aforementioned list of polymers is not exhaustive, and a variety of other synthetic hydrophilic polymers may be used, as will be appreciated by those skilled in the art. The synthetic crosslinkable hydrophilic polymer may be a homopolymer, a block copolymer, a random copolymer, or a graft copolymer. In addition, the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer. The polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments are those that degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ. Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc. Larger biodegradable "blocks" will generally be composed of oligomeric or polymeric segments incorporated within the hydrophilic polymer. Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like.
Other suitable synthetic crosslinkable hydrophilic polymers include chemically synthesized polypeptides, particularly polynucleophilic polypeptides that have been synthesized to incorporate amino acids containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine). Poly(lysine), a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred. Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000. Poly(lysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000, more preferably within the range of about 5,000 to about 100,000, and most preferably, within the range of about 8,000 to about 15,000. Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.).
The synthetic crosslinkable hydrophilic polymer may be a homopolymer, a block copolymer, a random copolymer, or a graft copolymer. In addition, the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer. The polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments are those that degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ. Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc. Larger biodegradable "blocks" will generally be composed of oligomeric or polymeric segments incorporated within the hydrophilic polymer. Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like.
Although a variety of different synthetic crosslinkable hydrophilic polymers can be used in the present compositions, as indicated above, preferred synthetic crosslinkable hydrophilic polymers are polyethylene glycol (PEG) and polyglycerol (PG), particularly highly branched polyglycerol. Various forms of PEG are extensively used in the modification of biologically active molecules because PEG lacks toxicity, antigenicity, and immunogenicity (i.e., is biocompatible), can be formulated so as to have a wide range of solubilities, and do not typically interfere with the enzymatic activities and/or conformations of peptides. A particularly preferred synthetic crosslinkable hydrophilic polymer for certain applications is a polyethylene glycol (PEG) having a molecular weight within the range of about 100 to about 100,000 mol. wt., although for highly branched PEG, far higher molecular weight polymers can be employed - up to 1 ,000,000 or more - providing that biodegradable sites are incorporated ensuring that all degradation products will have a molecular weight of less than about 30,000. For most PEGs, however, the preferred molecular weight is about 1 ,000 to about 20,000 mol. wt., more preferably within the range of about 7,500 to about 20,000 mol. wt. Most preferably, the polyethylene glycol has a molecular weight of approximately 10,000 mol. wt. Naturally occurring crosslinkable hydrophilic polymers include, but are not limited to: proteins such as collagen, fibronectin, albumins, globulins, fibrinogen, and fibrin, with collagen particularly preferred; carboxylated polysaccharides such as polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives. Collagen and glycosaminoglycans are examples of naturally occurring hydrophilic polymers for use herein, with methylated collagen being a preferred hydrophilic polymer.
Any of the hydrophilic polymers herein must contain, or be activated to contain, functional groups, i.e., nucleophilic or electrophilic groups, which enable crosslinking. Activation of PEG is discussed below; it is to be understood, however, that the following discussion is for purposes of illustration and analogous techniques may be employed with other polymers.
With respect to PEG, first of all, various functionalized polyethylene glycols have been used effectively in fields such as protein modification (see Abuchowski et al., Enzymes as Drugs, John Wiley & Sons: New York, N.Y. (1981) pp. 367-383; and Dreborg et al., Crit. Rev. Therap. Drug Carrier Syst. (1990) 6:315), peptide chemistry (see Mutter et al., The Peptides, Academic: New York, N.Y. 2:285-332; and Zalipsky et al., Int. J. Peptide Protein Res. (1987) 30:740), and the synthesis of polymeric drugs (see Zalipsky et al., Eur. Polym. J. (1983) 19:1177; and Ouchi et al., J. Macromol. Sci. Chem. (1987) A24:1011). Activated forms of PEG, including multifunctionally activated PEG, are commercially available, and are also easily prepared using known methods. For example, see Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, NY (1992); and Shearwater Polymers, Inc. Catalog, Polyethylene Glycol Derivatives, Huntsville, Alabama (1997-1998). Structures for some specific, tetrafunctionally activated forms of PEG are shown in FIGS. 1 to 10 of U.S. Patent 5,874,500, as are generalized reaction products obtained by reacting the activated PEGs with multi-amino PEGs, i.e., a PEG with two or more primary amino groups. The activated PEGs illustrated have a pentaerythritol (2,2-bis(hydroxymethyl)-1 ,3-propanediol) core. Such activated PEGs, as will be appreciated by those in the art, are readily prepared by conversion of the exposed hydroxyl groups in the PEGylated polyol (i.e., the terminal hydroxyl groups on the PEG chains) to carboxylic acid groups (typically by reaction with an anhydride in the presence of a nitrogenous base), followed by esterification with N-hydroxysuccinimide, N- hydroxysulfosuccinimide, or the like, to give the polyfunctionally activated PEG.
Hydrophobic Polymers:
The crosslinkable compositions of the invention can also include hydrophobic polymers, although for most uses hydrophilic polymers are preferred. Polylactic acid and polyglycolic acid are examples of two hydrophobic polymers that can be used. With other hydrophobic polymers, only short-chain oligomers should be used, containing at most about 14 carbon atoms, to avoid solubility-related problems during reaction.
Low Molecular Weight Components: As indicated above, the molecular core of one or more of the crosslinkable components can also be a low molecular weight compound, i.e., a C2-Cu hydrocarbyl group containing zero to 2 heteroatoms selected from N, O, S and combinations thereof. Such a molecular core can be substituted with nucleophilic groups or with electrophilic groups. When the low molecular weight molecular core is substituted with primary amino groups, the component may be, for example, ethylenediamine (H2N-CH2CH2-NH2), tetramethylenediamine (H2N-(CH4)-NH2), pentamethylenediamine (cadaverine) (H2N-(CH5)-NH2), hexamethylenediamine (H2N-(CHe)-NH2), bis(2-aminoethyl)amine (HN-[CH2CH2-NH2]2), or tris(2- aminoethyl)amine (N-[CH2CH2-NH2J3).
Low molecular weight diols and polyols include trimethylolpropane, di(trimethylol propane), pentaerythritol, and diglycerol, all of which require activation with a base in order to facilitate their reaction as nucleophiles. Such diols and polyols may also be functionalized to provide di- and poly-carboxylic acids, functional groups that are, as noted earlier herein, also useful as nucleophiles under certain conditions. Polyacids for use in the present compositions include, without limitation, trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)-based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid), all of which are commercially available and/or readily synthesized using known techniques.
Low molecular weight di- and poly-electrophiles include, for example, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS3), dithiobis(succinimidylpropionate) (DSP), bis(2-succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'-dithiobis(sulfosuccinimidylpropionate (DTSPP), and their analogs and derivatives. The aforementioned compounds are commercially available from Pierce (Rockford, III.). Such di- and poly- electrophiles can also be synthesized from di- and polyacids, for example by reaction with an appropriate molar amount of N-hydroxysuccinimide in the presence of DCC. Polyols such as trimethylolpropane and di(trimethylol propane) can be converted to carboxylic acid form using various known techniques, then further derivatized by reaction with NHS in the presence of DCC to produce trifunctionally and tetrafunctionally activated polymers.
Delivery Systems:
Suitable delivery systems for the homogeneous dry powder composition (containing at least two crosslinkable polymers) and the two buffer solutions may involve a multi-compartment spray device, where one or more compartments contains the powder and one or more compartments contain the buffer solutions needed to provide for the aqueous environment, so that the composition is exposed to the aqueous environment as it leaves the compartment. Many devices that are adapted for delivery of multi-component tissue sealants/hemostatic agents are well known in the art and can also be used in the practice of the present invention. Alternatively, the composition can be delivered using any type of controllable extrusion system, or it can be delivered manually in the form of a dry powder, and exposed to the aqueous environment at the site of administration.
The homogeneous dry powder composition and the two buffer solutions may be conveniently formed under aseptic conditions by placing each of the three ingredients (dry powder, acidic buffer solution and basic buffer solution) into separate syringe barrels. For example, the composition, first buffer solution and second buffer solution can be housed separately in a multiple-compartment syringe system having a multiple barrels, a mixing head, and an exit orifice. The first buffer solution can be added to the barrel housing the composition to dissolve the composition and form a homogeneous solution, which is then extruded into the mixing head. The second buffer solution can be simultaneously extruded into the mixing head. Finally, the resulting composition can then be extruded through the orifice onto a surface. For example, the syringe barrels holding the dry powder and the basic buffer may be part of a dual-syringe system, e.g., a double barrel syringe as described in U.S. Patent 4,359,049 to Redl et al. In this embodiment, the acid buffer can be added to the syringe barrel that also holds the dry powder, so as to produce the homogeneous solution. In other words, the acid buffer may be added (e.g., injected) into the syringe barrel holding the dry powder to thereby produce a homogeneous solution of the first and second components. This homogeneous solution can then be extruded into a mixing head, while the basic buffer is simultaneously extruded into the mixing head. Within the mixing head, the homogeneous solution and the basic buffer are mixed together to thereby form a reactive mixture. Thereafter, the reactive mixture is extruded through an orifice and onto a surface (e.g., tissue), where a film is formed, which can function as a sealant or a barrier, or the like. The reactive mixture begins forming a three-dimensional matrix immediately upon being formed by the mixing of the homogeneous solution and the basic buffer in the mixing head. Accordingly, the reactive mixture is preferably extruded from the mixing head onto the tissue very quickly after it is formed so that the three-dimensional matrix forms on, and is able to adhere to, the tissue.
Other systems for combining two reactive liquids are well known in the art, and include the systems described in U.S. Patent Nos. 6,454,786 to Holm et al.; 6,461 ,325 to Delmotte et al.; 5,585,007 to Antanavich et al.; 5,116,315 to Capozzi et al.; and 4,631,055 to Redl et al.
Storage and Handling:
Because crosslinkable components containing electrophilic groups react with water, the electrophilic component or components are generally stored and used in sterile, dry form to prevent hydrolysis. Processes for preparing synthetic hydrophilic polymers containing multiple electrophilic groups in sterile, dry form are set forth in commonly assigned U.S. Patent No.
5,643,464 to Rhee et al. For example, the dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or, preferably, e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates.
Components containing multiple nucleophilic groups are generally not water-reactive and can therefore be stored either dry or in aqueous solution.
If stored as a dry, particulate, solid, the various components of the crosslinkable composition may be blended and stored in a single container. Admixture of all components with water, saline, or other aqueous media should not occur until immediately prior to use.
In an alternative embodiment, the crosslinking components can be mixed together in a single aqueous medium in which they are both unreactive, i.e., such as in a low pH buffer. Thereafter, they can be sprayed onto the targeted tissue site along with a high pH buffer, after which they will rapidly react and form a gel.
Suitable liquid media for storage of crosslinkable compositions include aqueous buffer solutions such as monobasic sodium phosphate/dibasic sodium phosphate, sodium carbonate/sodium bicarbonate, glutamate or acetate, at a concentration of 0.5 to 300 mM. In general, a sulfhydryl-reactive component such as PEG substituted with maleimido groups or succinimidyl esters is prepared in water or a dilute buffer, with a pH of between around 5 to 6. Buffers with pKs between about 8 and 10.5 for preparing a polysulfhydryl component such as sulfhydryl-PEG are useful to achieve fast gelation time of compositions containing mixtures of sulfhydryl-PEG and SG-PEG. These include carbonate, borate and AMPSO (3-[(1 ,1-dimethyl-2- hydroxyethyl)amino]2-hydroxy-propane-sulfonic acid). In contrast, using a combination of maleimidyl PEG and sulfhydryl-PEG, a pH of around 5 to 9 is preferred for the liquid medium used to prepare the sulfhydryl PEG.
Collagen + Fibrinogen and/or Thrombin (e.g., Costasis)
In yet another aspect, the polymer composition may include collagen in combination with fibrinogen and/or thrombin. (See, e.g., U.S. Patent Nos. 5,290,552; 6,096,309; and 5,997,811). For example, an aqueous composition may include a fibrinogen and FXIII, particularly plasma, collagen in an amount sufficient to thicken the composition, thrombin in an amount sufficient to catalyze polymerization of fibrinogen present in the composition, and Ca2+ and, optionally, an antifibrinolytic agent in amount sufficient to retard degradation of the resulting adhesive clot. The composition may be formulated as a two-part composition that may be mixed together just prior to use, in which fibrinogen/FXIII and collagen constitute the first component, and thrombin together with an antifibrinolytic agent, and Ca2+ constitute the second component.
Plasma, which provides a source of fibrinogen, may be obtained from the patient for which the composition is to be delivered. The plasma can be used "as is" after standard preparation which includes centrifuging out cellular components of blood. Alternatively, the plasma can be further processed to concentrate the fibrinogen to prepare a plasma cryoprecipitate. The plasma cryoprecipitate can be prepared by freezing the plasma for at least about an hour at about -20°C, and then storing the frozen plasma overnight at about 4°C to slowly thaw. The thawed plasma is centrifuged and the plasma cryoprecipitate is harvested by removing approximately four-fifths of the plasma to provide a cryoprecipitate comprising the remaining one-fifth of the plasma. Other fibrinogen/FXIII preparations may be used, such as cryoprecipitate, patient autologous fibrin sealant, fibrinogen analogs or other single donor or commercial fibrin sealant materials. Approximately 0.5 ml to about 1.0 ml of either the plasma or the plasma-cryoprecipitate provides about 1 to 2 ml of adhesive composition which is sufficient for use in middle ear surgery. Other plasma proteins (e.g., albumin, plasminogen, von Willebrands factor, Factor VIII, etc.) may or may not be present in the fibrinogen/FXII separation due to wide variations in the formulations and methods to derive them.
Collagen, preferably hypoallergenic collagen, is present in the composition in an amount sufficient to thicken the composition and augment the cohesive properties of the preparation. The collagen may be atelopeptide collagen or telopeptide collagen, e.g., native collagen. In addition to thickening the composition, the collagen augments the fibrin by acting as a macromolecular lattice work or scaffold to which the fibrin network adsorbs. This gives more strength and durability to the resulting glue clot with a relatively low concentration of fibrinogen in comparison to the various concentrated autogenous fibrinogen glue formulations (i.e., AFGs).
The form of collagen which is employed may be described as at least "near native" in its structural characteristics. It may be further characterized as resulting in insoluble fibers at a pH above 5; unless crosslinked or as part of a complex composition, e.g., bone, it will generally consist of a minor amount by weight of fibers with diameters greater than 50 nm, usually from about 1 to 25 volume % and there will be substantially little, if any, change in the helical structure of the fibrils. In addition, the collagen composition must be able to enhance gelation in the surgical adhesion composition.
A number of commercially available collagen preparations may be used. ZYDERM Collagen Implant (ZCI) has a fibrillar diameter distribution consisting of 5 to 10 nm diameter fibers at 90% volume content and the remaining 10 % with greater than about 50 nm diameter fibers. ZCI is available as a fibrillar slurry and solution in phosphate buffered isotonic saline, pH 7.2, and is injectable with fine gauge needles. As distinct from ZCI, cross-linked collagen available as ZYPLAST may be employed. ZYPLAST is essentially an exogenously crosslinked (glutaraldehyde) version of ZCI. The material has a somewhat higher content of greater than about 50 nm diameter fibrils and remains insoluble over a wide pH range. Crosslinking has the effect of mimicking in vivo endogenous crosslinking found in many tissues. Thrombin acts as a catalyst for fibrinogen to provide fibrin, an insoluble polymer and is present in the composition in an amount sufficient to catalyze polymerization of fibrinogen present in the patient plasma. Thrombin also activates FXIII, a plasma protein that catalyzes covalent crosslinks in fibrin, rendering the resultant clot insoluble. Usually the thrombin is present in the adhesive composition in concentration of from about 0.01 to about 1000 or greater NIH units (NIHu) of activity, usually about i to about 500 NIHu, most usually about 200 to about 500 NIHu. The thrombin can be from a variety of host animal sources, conveniently bovine. Thrombin is commercially available from a variety of sources including Parke-Davis, usually lyophilized with buffer salts and stabilizers in vials which provide thrombin activity ranging from about 1000 NIHu to 10,000 NIHu. The thrombin is usually prepared by reconstituting the powder by the addition of either sterile distilled water or isotonic saline. Alternately, thrombin analogs or reptile-sourced coagulants may be used.
The composition may additionally comprise an effective amount of an antifibrinolytic agent to enhance the integrity of the glue clot as the healing processes occur. A number of antifibrinolytic agents are well known and include aprotinin, C1 -esterase inhibitor and ε-amino-n-caproic acid (EACA). ε- amino-n-caproic acid, the only antifibrinolytic agent approved by the FDA, is effective at a concentration of from about 5 mg/ml to about 40 mg/ml of the final adhesive composition, more usually from about 20 to about 30 mg/ml. EACA is commercially available as a solution having a concentration of about 250 mg/ml. Conveniently, the commercial solution is diluted with distilled water to provide a solution of the desired concentration. That solution is desirably used to reconstitute lyophilized thrombin to the desired thrombin concentration.
Other examples of in situ forming materials based on the crosslinking of proteins are described, e.g., in U.S. Patent Nos. RE38158;
4,839,345; 5,514,379, 5,583,114; 6,458,147; 6,371 ,975; 5,290,552; 6,096,309; U.S. Patent Application Publication Nos 2002/0161399; 2001/0018598 and PCT Publication Nos. WO 03/090683; WO 01/45761 ; WO 99/66964 and WO 96/03159).
Self-Reactive Compounds
In one aspect, the therapeutic agent is released from a crosslinked matrix formed, at least in part, from a self-reactive compound. As used herein, a self-reactive compound comprises a core substituted with a minimum of three reactive groups. The reactive groups may be directed attached to the core of the compound, or the reactive groups may be indirectly attached to the compound's core, e.g., the reactive groups are joined to the core through one or more linking groups.
Each of the three reactive groups that are necessarily present in a self-reactive compound can undergo a bond-forming reaction with at least one of the remaining two reactive groups. For clarity it is mentioned that when these compounds react to form a crosslinked matrix, it will most often happen that reactive groups on one compound will reactive with reactive groups on another compound. That is, the term "self-reactive" is not intended to mean that each self-reactive compound necessarily reacts with itself, but rather that when a plurality of identical self-reactive compounds are in combination and undergo a crosslinking reaction, then these compounds will react with one another to form the matrix. The compounds are "self-reactive" in the sense that they can react with other compounds having the identical chemical structure as themselves. The self-reactive compound comprises at least four components: a core and three reactive groups. In one embodiment, the self-reactive compound can be characterized by the formula (I), where R is the core, the reactive groups are represented by X1, X2 and X3, and a linker (L) is optionally present between the core and a functional group.
Figure imgf000381_0001
The core R is a polyvalent moiety having attachment to at least three groups (i.e., it is at least trivalent) and may be, or may contain, for example, a hydrophilic polymer, a hydrophobic polymer, an amphiphilic polymer, a C2-i4 hydrocarbyl, or a C2-i4 hydrocarbyl which is heteroatom- containing. The linking groups L1, L2, and L3 may be the same or different. The designators p, q and r are either 0 (when no linker is present) or 1 (when a linker is present). The reactive groups X1, X2 and X3 may be the same or different. Each of these reactive groups reacts with at least one other reactive group to form a three-dimensional matrix. Therefore X1 can react with X2 and/or X3, X2 can react with X1 and/or X3, X3 can react with X1 and/or X2 and so forth. A trivalent core will be directly or indirectly bonded to three functional groups, a tetravalent core will be directly or indirectly bonded to four functional groups, etc.
Each side chain typically has one reactive group. However, the invention also encompasses self-reactive compounds where the side chains contain more than one reactive group. Thus, in another embodiment of the invention, the self-reactive compound has the formula (II): [X' -(L4)a -Y' - (L5)b ] c R1
where: a and b are integers from 0-1 ; c is an integer from 3-12; R1 is selected from hydrophilic polymers, hydrophobic polymers, amphiphilic polymers, C2-14 hydrocarbyls, and heteroatom-containing C2-M hydrocarbyls; X' and Y' are reactive groups and can be the same or different; and L4 and L5 are linking groups. Each reactive group inter-reacts with the other reactive group to form a three-dimensional matrix. The compound is essentially non-reactive in an initial environment but is rendered reactive upon exposure to a modification in the initial environment that provides a modified environment such that a plurality of the self-reactive compounds inter-react in the modified environment to form a three-dimensional matrix. In one preferred embodiment, R is a hydrophilic polymer. In another preferred embodiment, X1 is a nucleophilic group and Y' is an electrophilic group.
The following self-reactive compound is one example of a compound of formula (II):
Figure imgf000382_0001
where R4 has the formula:
Figure imgf000382_0002
Thus, in formula (II), a and b are 1 ; c is 4; the core R' is the hydrophilic polymer, tetrafunctionally activated polyethylene glycol, (C(CH2-O- )4; X1 is the electrophilic reactive group, succinimidyl; Y1 is the nucleophilic reactive group -CH-NH2; L4 is -C(O)-O-; and L5 is -(CH2- C H2-O-C H2)x-C H2-O-
C(O)-(CHz)2-.
The self-reactive compounds of the invention are readily synthesized by techniques that are well known in the art. An exemplary synthesis is set forth below:
Figure imgf000383_0001
Mitsunobo or DCC
Figure imgf000383_0002
H2, Pd/C
Figure imgf000384_0001
The reactive groups are selected so that the compound is essentially non-reactive in an initial environment. Upon exposure to a specific modification in the initial environment, providing a modified environment, the compound is rendered reactive and a plurality of self-reactive compounds are then able to inter-react in the modified environment to form a three-dimensional matrix. Examples of modification in the initial environment are detailed below, but include the addition of an aqueous medium, a change in pH, exposure to ultraviolet radiation, a change in temperature, or contact with a redox initiator. The core and reactive groups can also be selected so as to provide a compound that has one of more of the following features: are biocompatible, are non-immunogenic, and do not leave any toxic, inflammatory or immunogenic reaction products at the site of administration. Similarly, the core and reactive groups can also be selected so as to provide a resulting matrix that has one or more of these features.
In one embodiment of the invention, substantially immediately or immediately upon exposure to the modified environment, the self-reactive compounds inter-react form a three-dimensional matrix. The term "substantially immediately" is intended to mean within less than five minutes, preferably within less than two minutes, and the term "immediately" is intended to mean within less than one minute, preferably within less than 30 seconds.
In one embodiment, the self-reactive compound and resulting matrix are not subject to enzymatic cleavage by matrix metalloproteinases such as collagenase, and are therefore not readily degradable in vivo. Further, the self-reactive compound may be readily tailored, in terms of the selection and quantity of each component, to enhance certain properties, e.g., compression strength, swellability, tack, hydrophilicity, optical clarity, and the like.
In one preferred embodiment, R is a hydrophilic polymer. In another preferred embodiment, X is a nucleophilic group, Y is an electrophilic group and Z is either an electrophilic or a nucleophilic group. Additional embodiments are detailed below.
A higher degree of inter-reaction, e.g., crosslinking, may be useful when a less swellable matrix is desired or increased compressive strength is desired. In those embodiments, it may be desirable to have n be an integer from 2-12. In addition, when a plurality of self-reactive compounds are utilized, the compounds may be the same or different.
Reactive Groups
Prior to use, the self-reactive compound is stored in an initial environment that insures that the compound remain essentially non-reactive until use. Upon modification of this environment, the compound is rendered reactive and a plurality of compounds will then inter-react to form the desired matrix. The initial environment, as well as the modified environment, is thus determined by the nature of the reactive groups involved. The number of reactive groups can be the same or different.
However, in one embodiment of the invention, the number of reactive groups is approximately equal. As used in this context, the term "approximately" refers to a 2:1 to 1 :2 ratio of moles of one reactive group to moles of a different reactive groups. A 1 :1 :1 molar ratio of reactive groups is generally preferred. In general, the concentration of the self-reactive compounds in the modified environment, when liquid in nature, will be in the range of about 1 to 50 wt%, generally about 2 to 40 wt%. The preferred concentration of the compound in the liquid will depend on a number of factors, including the type of compound (i.e., type of molecular core and reactive groups), its molecular weight, and the end use of the resulting three-dimensional matrix. For example, use of higher concentrations of the compounds, or using highly functionalized compounds, will result in the formation of a more tightly crosslinked network, producing a suffer, more robust gel. As such, compositions intended for use in tissue augmentation will generally employ concentrations of self-reactive compounds that fall toward the higher end of the preferred concentration range. Compositions intended for use as bioadhesives or in adhesion prevention do not need to be as firm and may therefore contain lower concentrations of the self-reactive compounds.
Electrophilic and Nucleophilic Reactive Groups In one embodiment of the invention, the reactive groups are electrophilic and nucleophilic groups, which undergo a nucleophilic substitution reaction, a nucleophilic addition reaction, or both. The term "electrophilic" refers to a reactive group that is susceptible to nucleophilic attack, i.e., susceptible to reaction with an incoming nucleophilic group. Electrophilic groups herein are positively charged or electron-deficient, typically electron- deficient. The term "nucleophilic" refers to a reactive group that is electron rich, has an unshared pair of electrons acting as a reactive site, and reacts with a positively charged or electron-deficient site. For such reactive groups, the modification in the initial environment comprises the addition of an aqueous medium and/or a change in pH.
In one embodiment of the invention, X1 (also referred to herein as X) can be a nucleophilic group and X2 (also referred to herein as Y) can be an electrophilic group or vice versa, and X3 (also referred to herein as Z) can be either an electrophilic or a nucleophilic group. X may be virtually any nucleophilic group, so long as reaction can occur with the electrophilic group Y and also with Z, when Z is electrophilic (ZEL). Analogously, Y may be virtually any electrophilic group, so long as reaction can take place with X and also with Z when Z is nucleophilic (ZNu)- The only limitation is a practical one, in that reaction between X and Y, and X and ZEL, or Y and ZNU should be fairly rapid and take place automatically upon admixture with an aqueous medium, without need for heat or potentially toxic or non-biodegradable reaction catalysts or other chemical reagents. It is also preferred although not essential that reaction occur without need for ultraviolet or other radiation. In one embodiment, the reactions between X and Y, and between either X and ZEL or Y and ZNU, are complete in under 60 minutes, preferably under 30 minutes. Most preferably, the reaction occurs in about 5 to 15 minutes or less.
Examples of nucleophilic groups suitable as X or FΠNU include, but are not limited to: -NH2, -NHR1, -N(R1)2, -SH, -OH, -COOH, -C6H4-OH, -H, -PH2,
-PHR1, -P(R1)2, -NH-NH2, -CO-NH-NH2, -C5H4N, etc. wherein R1 is a hydrocarbyl group and each R1 may be the same or different. R1 is typically alkyl or monocyclic aryl, preferably alkyl, and most preferably lower alkyl. Organometallic moieties are also useful nucleophilic groups for the purposes of the invention, particularly those that act as carbanion donors. Examples of organometallic moieties include: Grignard functionalities -R2MgHaI wherein R2 is a carbon atom (substituted or unsubstituted), and Hal is halo, typically bromo, iodo or chloro, preferably bromo; and lithium-containing functionalities, typically alkyllithium groups; sodium-containing functionalities.
It will be appreciated by those of ordinary skill in the art that certain nucleophilic groups must be activated with a base so as to be capable of reaction with an electrophilic group. For example, when there are nucleophilic sulfhydryl and hydroxyl groups in the self-reactive compound, the compound must be admixed with an aqueous base in order to remove a proton and provide an -S" or -O" species to enable reaction with the electrophilic group. Unless it is desirable for the base to participate in the reaction, a non- nucleophilic base is preferred. In some embodiments, the base may be present as a component of a buffer solution. Suitable bases and corresponding crosslinking reactions are described herein.
The selection of electrophilic groups provided on the self-reactive compound, must be made so that reaction is possible with the specific nucleophilic groups. Thus, when the X reactive groups are amino groups, the Y and any ZEL groups are selected so as to react with amino groups. Analogously, when the X reactive groups are sulfhydryl moieties, the corresponding electrophilic groups are sulfhydryl-reactive groups, and the like. In general, examples of electrophilic groups suitable as Y or ZEL include, but are not limited to, -CO-CI, -(CO)-O-(CO)-R (where R is an alkyl group), -CH=CH-CH=O and -CH=CH-C(CH3)=O, halo, -N=C=O, -N=C=S, -SO2CH=CH2, -0(CO)-C=CH2, -O(CO)-C(CH3)=CH2l -S-S-(C5H4N), -O(CO)-C(CH2CH3)=CH2, -CH=CH-C=NH, -COOH, -(CO)O-N(COCH2)2, -CHO, -(CO)O-N(COCHs)2-S(O)2OH, and -N(COCH)2.
When X is amino (generally although not necessarily primary amino), the electrophilic groups present on Y and ZEL are amine-reactive groups. Exemplary amine-reactive groups include, by way of example and not limitation, the following groups, or radicals thereof: (1) carboxylic acid esters, including cyclic esters and "activated" esters; (2) acid chloride groups (-CO-CI); (3) anhydrides (-(CO)-O-(CO)-R, where R is an alkyl group); (4) ketones and aldehydes, including α,β-unsaturated aldehydes and ketones such as -CH=CH-CH=O and -CH=CH-C(CH3)=O; (5) halo groups; (6) isocyanate group (-N=C=O); (7) thioisocyanato group (-N=C=S); (8) epoxides; (9) activated hydroxyl groups (e.g., activated with conventional activating agents such as carbonyldiimidazole or sulfonyl chloride); and (10) olefins, including conjugated olefins, such as ethenesulfonyl (-SO2CH=CH2) and analogous functional groups, including acrylate (-0(CO)-C=CH2), methacrylate (-O(CO)-C(CH3)=CH2), ethyl acrylate (-O(CO)-C(CH2CH3)=CH2), and ethyleneimino (-CH=CH-C=NH). In one embodiment the amine-reactive groups contain an electrophilically reactive carbonyl group susceptible to nucleophilic attack by a primary or secondary amine, for example the carboxylic acid esters and aldehydes noted above, as well as carboxyl groups (-COOH).
Since a carboxylic acid group perse is not susceptible to reaction with a nucleophilic amine, components containing carboxylic acid groups must be activated so as to be amine-reactive. Activation may be accomplished in a variety of ways, but often involves reaction with a suitable hydroxyl-containing compound in the presence of a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU). For example, a carboxylic acid can be reacted with an alkoxy-substituted N-hydroxy- succinimide or N-hydroxysulfosuccinimide in the presence of DCC to form reactive electrophilic groups, the N-hydroxysuccinimide ester and the N- hydroxysulfosuccinimide ester, respectively. Carboxylic acids may also be activated by reaction with an acyl halide such as an acyl chloride (e.g., acetyl chloride), to provide a reactive anhydride group. In a further example, a carboxylic acid may be converted to an acid chloride group using, e.g., thionyl chloride or an acyl chloride capable of an exchange reaction. Specific reagents and procedures used to carry out such activation reactions will be known to those of ordinary skill in the art and are described in the pertinent texts and literature. Accordingly, in one embodiment, the amine-reactive groups are selected from succinimidyl ester (-O(CO)-N(COCH2)2), sulfosuccinimidyl ester (-O(CO)-N(COCH2)2-S(O)2OH), maleimido (-N(COCH)2), epoxy, isocyanato, thioisocyanato, and ethenesulfonyl. Analogously, when X is sulfhydryl, the electrophilic groups present on Y and ZEL are groups that react with a sulfhydryl moiety. Such reactive groups include those that form thioester linkages upon reaction with a sulfhydryl group, such as those described in WO 00/62827 to Wallace et al. As explained in detail therein, sulfhydryl reactive groups include, but are not limited to: mixed anhydrides; ester derivatives of phosphorus; ester derivatives of p-nitrophenol, p-nitrothiophenol and pentafluorophenol; esters of substituted hydroxylamines, including N-hydroxyphthalimide esters, N-hydroxysuccinimide esters, N- hydroxysulfosuccinimide esters, and N-hydroxyglutarimide esters; esters of 1- hydroxybenzotriazole; 3-hydroxy-3,4-dihydro-benzotriazin-4-one; 3-hydroxy- 3,4-dihydro-quinazoline-4-one; carbonylimidazole derivatives; acid chlorides; ketenes; and isocyanates. With these sulfhydryl reactive groups, auxiliary reagents can also be used to facilitate bond formation, e.g., 1-ethyl-3-[3- dimethylaminopropyl]carbodiimide can be used to facilitate coupling of sulfhydryl groups to carboxyl-containing groups. In addition to the sulfhydryl reactive groups that form thioester linkages, various other sulfhydryl reactive functionalities can be utilized that form other types of linkages. For example, compounds that contain methyl imidate derivatives form imido-thioester linkages with sulfhydryl groups. Alternatively, sulfhydryl reactive groups can be employed that form disulfide bonds with sulfhydryl groups; such groups generally have the structure -S-S-Ar where Ar is a substituted or unsubstituted nitrogen-containing heteroaromatic moiety or a non-heterocyclic aromatic group substituted with an electron- withdrawing moiety, such that Ar may be, for example, 4-pyridinyl, o- nitrophenyl, m-nitrophenyl, p-nitrophenyl, 2,4-dinitrophenyl, 2-nitro-4-benzoic acid, 2-nitro-4-pyridinyl, etc. In such instances, auxiliary reagents, i.e., mild oxidizing agents such as hydrogen peroxide, can be used to facilitate disulfide bond formation.
Yet another class of sulfhydryl reactive groups forms thioether bonds with sulfhydryl groups. Such groups include, inter alia, maleimido, substituted maleimido, haloalkyl, epoxy, imino, and aziridino, as well as olefins (including conjugated olefins) such as ethenesulfonyl, etheneimino, acrylate, methacrylate, and α,β-unsaturated aldehydes and ketones.
When X is -OH, the electrophilic functional groups on the remaining component(s) must react with hydroxyl groups. The hydroxyl group may be activated as described above with respect to carboxylic acid groups, or it may react directly in the presence of base with a sufficiently reactive electrophilic group such as an epoxide group, an aziridine group, an acyl halide, an anhydride, and so forth.
When X is an organometallic nucleophilic group such as a Grignard functionality or an alkyllithium group, suitable electrophilic functional groups for reaction therewith are those containing carbonyl groups, including, by way of example, ketones and aldehydes.
It will also be appreciated that certain functional groups can react as nucleophilic or as electrophilic groups, depending on the selected reaction partner and/or the reaction conditions. For example, a carboxylic acid group can act as a nucleophilic group in the presence of a fairly strong base, but generally acts as an electrophilic group allowing nucleophilic attack at the carbonyl carbon and concomitant replacement of the hydroxyl group with the incoming nucleophilic group. These, as well as other embodiments are illustrated below, where the covalent linkages in the matrix that result upon covalent binding of specific nucleophilic reactive groups to specific electrophilic reactive groups on the self- reactive compound include, solely by way of example, the following Table: Table
Figure imgf000392_0001
Figure imgf000393_0001
For self-reactive compounds containing electrophilic and nucleophilic reactive groups, the initial environment typically can be dry and sterile. Since electrophilic groups react with water, storage in sterile, dry form will prevent hydrolysis. The dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or e- beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates. The modification of a dry initial environment will typically comprise the addition of an aqueous medium. In one embodiment, the initial environment can be an aqueous medium such as in a low pH buffer, i.e., having a pH less than about 6.0, in which both electrophilic and nucleophilic groups are non-reactive. Suitable liquid media for storage of such compounds include aqueous buffer solutions such as monobasic sodium phosphate/dibasic sodium phosphate, sodium carbonate/sodium bicarbonate, glutamate or acetate, at a concentration of 0.5 to 300 mM. Modification of an initial low pH aqueous environment will typically comprise increasing the pH to at least pH 7.0, more preferably increasing the pH to at least pH 9.5.
In another embodiment the modification of a dry initial environment comprises dissolving the self-reactive compound in a first buffer solution having a pH within the range of about 1.0 to 5.5 to form a homogeneous solution, and (ii) adding a second buffer solution having a pH within the range of about 6.0 to 11.0 to the homogeneous solution. The buffer solutions are aqueous and can be any pharmaceutically acceptable basic or acid composition. The term "buffer" is used in a general sense to refer to an acidic or basic aqueous solution, where the solution may or may not be functioning to provide a buffering effect (i.e., resistance to change in pH upon addition of acid or base) in the compositions of the present invention. For example, the self-reactive compound can be in the form of a homogeneous dry powder. This powder is then combined with a buffer solution having a pH within the range of about 1.0 to 5.5 to form a homogeneous acidic aqueous solution, and this solution is then combined with a buffer solution having a pH within the range of about 6.0 to 11.0 to form a reactive solution. For example, 0.375 grams of the dry powder can be combined with 0.75 grams of the acid buffer to provide, after mixing, a homogeneous solution, where this solution is combined with 1.1 grams of the basic buffer to provide a reactive mixture that substantially immediately forms a three-dimensional matrix.
Acidic buffer solutions having a pH within the range of about 1.0 to 5.5, include by way of illustration and not limitation, solutions of: citric acid, hydrochloric acid, phosphoric acid, sulfuric acid, AMPSO (3-[(1,1-dimethyl-2- hydroxyethyl)amino]2-hydroxy-propane-sulfonic acid), acetic acid, lactic acid, and combinations thereof. In a preferred embodiment, the acidic buffer solution, is a solution of citric acid, hydrochloric acid, phosphoric acid, sulfuric acid, and combinations thereof. Regardless of the precise acidifying agent, the acidic buffer preferably has a pH such that it retards the reactivity of the nucleophilic groups on the core. For example, a pH of 2.1 is generally sufficient to retard the nucleophilicity of thiol groups. A lower pH is typically preferred when the core contains amine groups as the nucleophilic groups. In general, the acidic buffer is an acidic solution that, when contacted with nucleophilic groups, renders those nucleophilic groups relatively non-nucleophilic.
An exemplary acidic buffer is a solution of hydrochloric acid, having a concentration of about 6.3 mM and a pH in the range of 2.1 to 2.3. This buffer may be prepared by combining concentrated hydrochloric acid with water, i.e., by diluting concentrated hydrochloric acid with water. Similarly, this buffer A may also be conveniently prepared by diluting 1.23 grams of concentrated hydrochloric acid to a volume of 2 liters, or diluting 1.84 grams of concentrated hydrochloric acid to a volume to 3 liters, or diluting 2.45 grams of concentrated hydrochloric acid to a volume of 4 liters, or diluting 3.07 grams concentrated hydrochloric acid to a volume of 5 liters, or diluting 3.68 grams of concentrated hydrochloric acid to a volume to 6 liters. For safety reasons, the concentrated acid is preferably added to water.
Basic buffer solutions having a pH within the range of about 6.0 to 11.0, include by way of illustration and not limitation, solutions of: glutamate, acetate, carbonate and carbonate salts (e.g., sodium carbonate, sodium carbonate monohydrate and sodium bicarbonate), borate, phosphate and phosphate salts (e.g., monobasic sodium phosphate monohydrate and dibasic sodium phosphate), and combinations thereof. In a preferred embodiment, the basic buffer solution is a solution of carbonate salts, phosphate salts, and combinations thereof. In general, the basic buffer is an aqueous solution that neutralizes the effect of the acidic buffer, when it is added to the homogeneous solution of the compound and first buffer, so that the nucleophilic groups on the core regain their nucleophilic character (that has been masked by the action of the acidic buffer), thus allowing the nucleophilic groups to inter-react with the electrophilic groups on the core.
An exemplary basic buffer is an aqueous solution of carbonate and phosphate salts. This buffer may be prepared by combining a base solution with a salt solution. The salt solution may be prepared by combining 34.7 g of monobasic sodium phosphate monohydrate, 49.3 g of sodium carbonate monohydrate, and sufficient water to provide a solution volume of 2 liter. Similarly, a 6 liter solution may be prepared by combining 104.0 g of monobasic sodium phosphate monohydrate, 147.94 g of sodium carbonate monohydrate, and sufficient water to provide 6 liter of the salt solution. The basic buffer may be prepared by combining 7.2 g of sodium hydroxide with 180.0 g of water. The basic buffer is typically prepared by adding the base solution as needed to the salt solution, ultimately to provide a mixture having the desired pH, e.g., a pH of 9.65 to 9.75.
In general, the basic species present in the basic buffer should be sufficiently basic to neutralize the acidity provided by the acidic buffer, but should not be so nucleophilic itself that it will react substantially with the electrophilic groups on the core. For this reason, relatively "soft" bases such as carbonate and phosphate are preferred in this embodiment of the invention.
To illustrate the preparation of a three-dimensional matrix of the present invention, one may combine an admixture of the self-reactive compound with a first, acidic, buffer (e.g., an acid solution, e.g., a dilute hydrochloric acid solution) to form a homogeneous solution. This homogeneous solution is mixed with a second, basic, buffer (e.g., a basic solution, e.g., an aqueous solution containing phosphate and carbonate salts) whereupon the reactive groups on the core of the self-reactive compound substantially immediately inter-react with one another to form a three- dimensional matrix.
Redox Reactive Groups
In one embodiment of the invention, the reactive groups are vinyl groups such as styrene derivatives, which undergo a radical polymerization upon initiation with a redox initiator. The term "redox" refers to a reactive group that is susceptible to oxidation-reduction activation. The term "vinyl" refers to a reactive group that is activated by a redox initiator, and forms a radical upon reaction. X, Y and Z can be the same or different vinyl groups, for example, methacrylic groups. For self-reactive compounds containing vinyl reactive groups, the initial environment typically will be an aqueous environment. The modification of the initial environment involves the addition of a redox initiator. Oxidative Coupling Reactive Groups
In one embodiment of the invention, the reactive groups undergo an oxidative coupling reaction. For example, X, Y and Z can be a halo group such as chloro, with an adjacent electron-withdrawing group on the halogen- bearing carbon (e.g., on the "L" linking group). Exemplary electron-withdrawing groups include nitro, aryl, and so forth.
For such reactive groups, the modification in the initial environment comprises a change in pH. For example, in the presence of a base such as KOH, the self-reactive compounds then undergo a de-hydro, chloro coupling reaction, forming a double bond between the carbon atoms, as illustrated below:
Figure imgf000397_0001
For self-reactive compounds containing oxidative coupling reactive groups, the initial environment typically can be can be dry and sterile, or a non-basic medium. The modification of the initial environment will typically comprise the addition of a base.
Photoinitiated Reactive Groups
In one embodiment of the invention, the reactive groups are photoinitiated groups. For such reactive groups, the modification in the initial environment comprises exposure to ultraviolet radiation.
In one embodiment of the invention, X can be an azide (-N3) group and Y can be an alkyl group such as -CH(CHs)2 or vice versa. Exposure to ultraviolet radiation will then form a bond between the groups to provide for the following linkage: -NH-C(CH3)2-CH2-. In another embodiment of the invention, X can be a benzophenone (-(C6H-I)-C(O)-(CeHs)) group and Y can be an alkyl group such as -CH(CHs)2 or vice versa. Exposure to ultraviolet radiation will then form a bond between the groups to provide for the following linkage:
Figure imgf000398_0001
For self-reactive compounds containing photoinitiated reactive groups, the initial environment typically will be in an ultraviolet radiation- shielded environment. This can be for example, storage within a container that is impermeable to ultraviolet radiation.
The modification of the initial environment will typically comprise exposure to ultraviolet radiation.
Temperature-sensitive Reactive Groups
In one embodiment of the invention, the reactive groups are temperature-sensitive groups, which undergo a thermochemical reaction. For such reactive groups, the modification in the initial environment thus comprises a change in temperature. The term "temperature-sensitive" refers to a reactive group that is chemically inert at one temperature or temperature range and reactive at a different temperature or temperature range.
In one embodiment of the invention, X, Y, and Z are the same or different vinyl groups. For self-reactive compounds containing reactive groups that are temperature-sensitive, the initial environment typically will be within the range of about 10 to 3O0C.
The modification of the initial environment will typically comprise changing the temperature to within the range of about 20 to 4O0C.
Linking Groups
The reactive groups may be directly attached to the core, or they may be indirectly attached through a linking group, with longer linking groups also termed "chain extenders." In the formula (I) shown above, the optional linker groups are represented by L1, L2, and L3, wherein the linking groups are present when p, q and r are equal to 1.
Suitable linking groups are well known in the art. See, for example, WO 97/22371 to Rhee et al. Linking groups are useful to avoid steric hindrance problems that can sometimes associated with the formation of direct linkages between molecules. Linking groups may additionally be used to link several self-reactive compounds together to make larger molecules. In one embodiment, a linking group can be used to alter the degradative properties of the compositions after administration and resultant gel formation. For example, linking groups can be used to promote hydrolysis, to discourage hydrolysis, or to provide a site for enzymatic degradation.
Examples of linking groups that provide hydrolyzable sites, include, inter alia: ester linkages; anhydride linkages, such as those obtained by incorporation of glutarate and succinate; ortho ester linkages; ortho carbonate linkages such as trimethylene carbonate; amide linkages; phosphoester linkages; α-hydroxy acid linkages, such as those obtained by incorporation of lactic acid and glycolic acid; lactone-based linkages, such as those obtained by incorporation of caprolactone, valerolactone, γ-butyrolactone and p-dioxanone; and amide linkages such as in a dimeric, oligomeric, or poly(amino acid) segment. Examples of non-degradable linking groups include succinimide, propionic acid and carboxymethylate linkages. See, for example, WO 99/07417 to Coury et al. Examples of enzymatically degradable linkages include Leu- Gly-Pro-Ala, which is degraded by collagenase; and Gly-Pro-Lys, which is degraded by plasmin.
Linking groups can also be included to enhance or suppress the reactivity of the various reactive groups. For example, electron-withdrawing groups within one or two carbons of a sulfhydryl group may be expected to diminish its effectiveness in coupling, due to a lowering of nucleophilicity. Carbon-carbon double bonds and carbonyl groups will also have such an effect. Conversely, electron-withdrawing groups adjacent to a carbonyl group (e.g., the reactive carbonyl of glutaryl-N-hydroxysuccinimidyl) may increase the reactivity of the carbonyl carbon with respect to an incoming nucleophilic group. By contrast, sterically bulky groups in the vicinity of a reactive group can be used to diminish reactivity and thus reduce the coupling rate as a result of steric hindrance.
By way of example, particular linking groups and corresponding formulas are indicated in the following Table:
Table
Figure imgf000400_0001
Figure imgf000401_0001
In the above Table, x is generally in the range of 1 to about 10; R2 is generally hydrocarbyl, typically alkyl or aryl, preferably alkyl, and most preferably lower alkyl; and R3 is hydrocarbylene, heteroatom-containing hydrocarbylene, substituted hydrocarbylene, or substituted heteroatom- containing hydrocarbylene) typically alkylene or arylene (again, optionally substituted and/or containing a heteroatom), preferably lower alkylene (e.g., methylene, ethylene, n-propylene, n-butylene, etc.), phenylene, or amidoalkylene (e.g., -(CO)-NH-CH2). Other general principles that should be considered with respect to linking groups are as follows. If a higher molecular weight self-reactive compound is to be used, it will preferably have biodegradable linkages as described above, so that fragments larger than 20,000 mol. wt. are not generated during resorption in the body. In addition, to promote water miscibiiity and/or solubility, it may be desired to add sufficient electric charge or hydrophilicity. Hydrophilic groups can be easily introduced using known chemical synthesis, so long as they do not give rise to unwanted swelling or an undesirable decrease in compressive strength. In particular, polyalkoxy segments may weaken gel strength. The Core
The "core" of each self-reactive compound is comprised of the molecular structure to which the reactive groups are bound. The molecular core can be a polymer, which includes synthetic polymers and naturally occurring polymers. In one embodiment, the core is a polymer containing repeating monomer units. The polymers can be hydrophilic, hydrophobic, or amphiphilic. The molecular core can also be a low molecular weight component such as a C2-I4 hydrocarbyl or a heteroatom-containing C2-i4 hydrocarbyl. The heteroatom-containing C2--I4 hydrocarbyl can have 1 or 2 heteroatoms selected from N, O and S. In a preferred embodiment, the self- reactive compound comprises a molecular core of a synthetic hydrophilic polymer.
Hydrophilic Polymers
As mentioned above, the term "hydrophilic polymer" as used herein refers to a polymer having an average molecular weight and composition that naturally renders, or is selected to render the polymer as a whole "hydrophilic." Preferred polymers are highly pure or are purified to a highly pure state such that the polymer is or is treated to become pharmaceutically pure. Most hydrophilic polymers can be rendered water soluble by incorporating a sufficient number of oxygen (or less frequently nitrogen) atoms available for forming hydrogen bonds in aqueous solutions.
Synthetic hydrophilic polymers may be homopolymers, block copolymers including di-block and tri-block copolymers, random copolymers, or graft copolymers. In addition, the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer. The polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments preferably degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ. Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc. Larger biodegradable "blocks" will generally be composed of oligomeric or polymeric segments incorporated within the hydrophilic polymer. Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like. Other biodegradable segments that may form part of the hydrophilic polymer core include polyesters such as polylactide, polyethers such as polyalkylene oxide, polyamides such as a protein, and polyurethanes. For example, the core of the self-reactive compound can be a diblock copolymer of tetrafunctionally activated polyethylene glycol and polylactide.
Synthetic hydrophilic polymers that are useful herein include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol (PEG) and poly(ethylene oxide)-poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (PG) and particularly highly branched polyglycerol, propylene glycol; poly(oxyalkylene)-substituted diols, and poly(oxyalkylene)-substituted polyols such as mono-, di- and tri- polyoxyethylated glycerol, mono- and di-polyoxyethylated propylene glycol, and mono- and di-polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; poly(acrylic acids) and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly( hyd roxyethylmeth acrylate) , poly(hyd roxyethylacrylate) , poly(methylalkylsulfoxide methacrylates), poly(methylalkylsulfoxide acrylates) and copolymers of any of the foregoing, and/or with additional acrylate species such as aminoethyl acrylate and mono-2-(acryloxy)-ethyl succinate; polymaleic acid; poly(acrylamides) such as polyacrylamide perse, poly(methacrylamide), poly(dimethylacrylamide), poly(N-isopropyl-acrylamide), and copolymers thereof; poly(olefinic alcohols) such as polyvinyl alcohols) and copolymers thereof; poly(N-vinyl lactams) such as polyvinyl pyrrolidones), poly(N-vinyl caproiactams), and copolymers thereof; polyoxazolines, including poly(methyloxazoline) and poly(ethyloxazoline); and polyvinylamines; as well as copolymers of any of the foregoing. It must be emphasized that the aforementioned list of polymers is not exhaustive, and a variety of other synthetic hydrophilic polymers may be used, as will be appreciated by those skilled in the art.
Those of ordinary skill in the art will appreciate that synthetic polymers such as polyethylene glycol cannot be prepared practically to have exact molecular weights, and that the term "molecular weight" as used herein refers to the weight average molecular weight of a number of molecules in any given sample, as commonly used in the art. Thus, a sample of PEG 2,000 might contain a statistical mixture of polymer molecules ranging in weight from, for example, 1,500 to 2,500 daltons with one molecule differing slightly from the next over a range. Specification of a range of molecular weights indicates that the average molecular weight may be any value between the limits specified, and may include molecules outside those limits. Thus, a molecular weight range of about 800 to about 20,000 indicates an average molecular weight of at least about 800, ranging up to about 20 kDa.
Other suitable synthetic hydrophilic polymers include chemically synthesized polypeptides, particularly polynucleophilic polypeptides that have been synthesized to incorporate amino acids containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine). Poly(lysine), a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred. Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000. Poly(lysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000, more preferably within the range of about 5,000 to about 100,000, and most preferably, within the range of about 8,000 to about 15,000. Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.). Although a variety of different synthetic hydrophilic polymers can be used in the present compounds, preferred synthetic hydrophilic polymers are PEG and PG, particularly highly branched PG. Various forms of PEG are extensively used in the modification of biologically active molecules because PEG lacks toxicity, antigenicity, and immunogenicity (i.e., is biocompatible), can be formulated so as to have a wide range of solubilities, and does not typically interfere with the enzymatic activities and/or conformations of peptides. A particularly preferred synthetic hydrophilic polymer for certain applications is a PEG having a molecular weight within the range of about 100 to about 100,000, although for highly branched PEG, far higher molecular weight polymers can be employed, up to 1 ,000,000 or more, providing that biodegradable sites are incorporated ensuring that all degradation products will have a molecular weight of less than about 30,000. For most PEGs, however, the preferred molecular weight is about 1 ,000 to about 20,000, more preferably within the range of about 7,500 to about 20,000. Most preferably, the polyethylene glycol has a molecular weight of approximately 10,000.
Naturally occurring hydrophilic polymers include, but are not limited to: proteins such as collagen, fibronectin, albumins, globulins, fibrinogen, fibrin and thrombin, with collagen particularly preferred; carboxylated polysaccharides such as polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives. Collagen and glycosaminoglycans are preferred naturally occurring hydrophilic polymers for use herein.
Unless otherwise specified, the term "collagen" as used herein refers to all forms of collagen, including those, which have been processed or otherwise modified. Thus, collagen from any source may be used in the compounds of the invention; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced. The preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. For example, U.S. Patent No. 5,428,022 to Palefsky et al. discloses methods of extracting and purifying collagen from the human placenta, and U.S. Patent No. 5,667,839 to Berg discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows. Non-transgenic, recombinant collagen expression in yeast and other cell lines) is described in U.S. Patent No. 6,413,742 to Olsen et al., 6,428,978 to Olsen et al., and 6,653,450 to Berg et al.
Collagen of any type, including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compounds of the invention, although type I is generally preferred. Either atelopeptide or telopeptide- containing collagen may be used; however, when collagen from a natural source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide- containing collagen.
Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the invention, although previously crosslinked collagen may be used.
Collagens for use in the present invention are generally, although not necessarily, in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml, preferably between about 30 mg/ml to about 90 mg/ml. Although intact collagen is preferred, denatured collagen, commonly known as gelatin, can also be used. Gelatin may have the added benefit of being degradable faster than collagen. Nonfibrillar collagen is generally preferred for use in compounds of the invention, although fibrillar collagens may also be used. The term "nonfibrillar collagen" refers to any modified or unmodified collagen material that is in substantially nonfibrillar form, i.e., molecular collagen that is not tightly associated with other collagen molecules so as to form fibers. Typically, a solution of nonfibrillar collagen is more transparent than is a solution of fibrillar collagen. Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen and methylated collagen, both of which can be prepared according to the methods described in U.S. Patent No. 4,164,559 to Miyata et al. Methylated collagen, which contains reactive amine groups, is a preferred nucleophile-containing component in the compositions of the present invention. In another aspect, methylated collagen is a component that is present in addition to first and second components in the matrix-forming reaction of the present invention. Methylated collagen is described in, for example, in U.S. Patent No. 5,614,587 to Rhee et al.
Collagens for use in the compositions of the present invention may start out in fibrillar form, then can be rendered nonfibrillar by the addition of one or more fiber disassembly agent. The fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above. Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids, inorganic salts, and carbohydrates, with biocompatible alcohols being particularly preferred. Preferred biocompatible alcohols include glycerol and propylene glycol. Non-biocompatible alcohols, such as ethanol, methanol, and isopropanol, are not preferred for use in the present invention, due to their potentially deleterious effects on the body of the patient receiving them. Preferred amino acids include arginine. Preferred inorganic salts include sodium chloride and potassium chloride. Although carbohydrates, such as various sugars including sucrose, may be used in the practice of the present invention, they are not as preferred as other types of fiber disassembly agents because they can have cytotoxic effects in vivo.
Fibrillar collagen is less preferred for use in the compounds of the invention. However, as disclosed in U.S. Patent No. 5,614,587 to Rhee et al., fibrillar collagen, or mixtures of nonfibrillar and fibrillar collagen, may be preferred for use in compounds intended for long-term persistence in vivo. Hydrophobic Polymers
The core of the self-reactive compound may also comprise a hydrophobic polymer, including low molecular weight polyfunctional species, although for most uses hydrophilic polymers are preferred. Generally, "hydrophobic polymers" herein contain a relatively small proportion of oxygen and/or nitrogen atoms. Preferred hydrophobic polymers for use in the invention generally have a carbon chain that is no longer than about 14 carbons. Polymers having carbon chains substantially longer than 14 carbons generally have very poor solubility in aqueous solutions and, as such, have very long reaction times when mixed with aqueous solutions of synthetic polymers containing, for example, multiple nucleophilic groups. Thus, use of short-chain oligomers can avoid solubility-related problems during reaction. Polylactic acid and polyglycolic acid are examples of two particularly suitable hydrophobic polymers.
Amphiphilic Polymers
Generally, amphiphilic polymers have a hydrophilic portion and a hydrophobic (or lipophilic) portion. The hydrophilic portion can be at one end of the core and the hydrophobic portion at the opposite end, or the hydrophilic and hydrophobic portions may be distributed randomly (random copolymer) or in the form of sequences or grafts (block copolymer) to form the amphiphilic polymer core of the self-reactive compound. The hydrophilic and hydrophobic portions may include any of the aforementioned hydrophilic and hydrophobic polymers.
Alternately, the amphiphilic polymer core can be a hydrophilic polymer that has been modified with hydrophobic moieties (e.g., alkylated PEG or a hydrophilic polymer modified with one or more fatty chains), or a hydrophobic polymer that has been modified with hydrophilic moieties (e.g., "PEGylated" phospholipids such as polyethylene glycolated phospholipids). Low Molecular Weight Components
As indicated above, the molecular core of the self-reactive compound can also be a low molecular weight compound, defined herein as being a C2-M hydrocarbyl or a heteroatom-containing C2-i4 hydrocarbyl, which contains 1 to 2 heteroatoms selected from N, O, S and combinations thereof. Such a molecular core can be substituted with any of the reactive groups described herein.
Alkanes are suitable C2-i4 hydrocarbyl molecular cores. Exemplary alkanes, for substituted with a nucleophilic primary amino group and a Y electrophilic group, include, ethyleneamine (H2N-CH2CH2-Y), tetramethyleneamine (H2N-(CH4)-Y), pentamethyleneamine (H2N-(CH5)-Y), and hexamethyleneamine (H2N-(C H6)-Y).
Low molecular weight diols and polyols are also suitable C2--I4 hydrocarbyls and include trimethylolpropane, di(trimethylol propane), pentaerythritol, and diglycerol. Polyacids are also suitable C2-I4 hydrocarbyls, and include trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)- based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid).
Low molecular weight di- and poly-electrophiles are suitable heteroatom-containing C2-I4 hydrocarbyl molecular cores. These include, for example, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS3), dithiobis(succinimidylpropionate) (DSP), bis(2-succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'-dithiobis(sulfosuccinimidylpropionate (DTSPP), and their analogs and derivatives. In one embodiment of the invention, the self-reactive compound of the invention comprises a low-molecular weight material core, with a plurality of acrylate moieties and a plurality of thiol groups.
Preparation
The self-reactive compounds are readily synthesized to contain a hydrophilic, hydrophobic or amphiphilic polymer core or a low molecular weight core, functionalized with the desired functional groups, i.e., nucleophilic and electrophilic groups, which enable crosslinking. For example, preparation of a self-reactive compound having a polyethylene glycol (PEG) core is discussed below. However, it is to be understood that the following discussion is for purposes of illustration and analogous techniques may be employed with other polymers.
With respect to PEG, first of all, various functionalized PEGs have been used effectively in fields such as protein modification (see Abuchowski et al., Enzymes as Drugs, John Wiley & Sons: New York, N.Y. (1981) pp. 367- 383; and Dreborg et al. (1990) Crit. Rev. Therap. Drug Carrier Syst 6:315), peptide chemistry (see Mutter et al., The Peptides, Academic: New York, N.Y. 2:285-332; and Zalipsky et al. (1987) Int. J. Peptide Protein Res. 30:740), and the synthesis of polymeric drugs (see Zalipsky et al. (1983) Eur. Polym. J. 19:1177; and Ouchi et al. (1987) J. Macromol. Sci. Chem. A24:1011). Functionalized forms of PEG, including multi-functionalized PEG, are commercially available, and are also easily prepared using known methods. For example, see Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, NY (1992). Multi-functionalized forms of PEG are of particular interest and include, PEG succinimidyl glutarate, PEG succinimidyl propionate, succinimidyl butylate, PEG succinimidyl acetate, PEG succinimidyl succinamide, PEG succinimidyl carbonate, PEG propionaldehyde, PEG glycidyl ether, PEG- isocyanate, and PEG-vinylsulfone. Many such forms of PEG are described in U.S. Patent No. 5,328,955 and 6,534,591 , both to Rhee et al. Similarly, various forms of multi-amino PEG are commercially available from sources such as PEG Shop, a division of SunBio of South Korea (www.sunbio.com), Nippon Oil and Fats (Yebisu Garden Place Tower, 20-3 Ebisu 4-chome, Shibuya-ku, Tokyo), Nektar Therapeutics (San Carlos, California, formerly Shearwater Polymers, Huntsville, Alabama) and from Huntsman's Performance Chemicals Group (Houston, Texas) under the name Jeffamine® polyoxyalkyleneamines. Multi-amino PEGs useful in the present invention include the Jeffamine diamines ("D" series) and triamines ("T" series), which contain two and three primary amino groups per molecule. Analogous poly(sulfhydryl) PEGs are also available from Nektar Therapeutics, e.g., in the form of pentaerythritol poly(ethylene glycol) ether tetra-sulfhydryl (molecular weight 10,000). These multi-functionalized forms of PEG can then be modified to include the other desired reactive groups.
Reaction with succinimidyl groups to convert terminal hydroxyl groups to reactive esters is one technique for preparing a core with electrophilic groups. This core can then be modified include nucleophilic groups such as primary amines, thiols, and hydroxyl groups. Other agents to convert hydroxyl groups include carbonyldiimidazole and sulfonyl chloride. However, as discussed herein, a wide variety of electrophilic groups may be advantageously employed for reaction with corresponding nucleophilic groups. Examples of such electrophilic groups include acid chloride groups; anhydrides, ketones, aldehydes, isocyanate, isothiocyanate, epoxides, and olefins, including conjugated olefins such as ethenesulfonyl (-SO2CH=CH2) and analogous functional groups.
Other in situ Crosslinking Materials
Numerous other types of in situ forming materials have been described which may be used in combination with an anti-scarring agent in accordance with the invention. The in situ forming material may be a biocompatible crosslinked polymer that is formed from water soluble precursors having electrophilic and nucleophilic groups capable of reacting and crosslinking in situ (see, e.g., U.S. Patent No. 6,566,406). The in situ forming material may be hydrogel that may be formed through a combination of physical and chemical crosslinking processes, where physical crosslinking is mediated by one or more natural or synthetic components that stabilize the hydrogel- forming precursor solution at a deposition site for a period of time sufficient for more resilient chemical crosslinks to form (see, e.g., U.S. Patent No. 6,818,018). The in situ forming material may be formed upon exposure to an aqueous fluid from a physiological environment from dry hydrogel precursors (see, e.g., U.S. Patent No. 6,703,047). The in situ forming material may be a hydrogel matrix that provides controlled release of relatively low molecular weight therapeutic species by first dispersing or dissolving the therapeutic species within relatively hydrophobic rate modifying agents to form a mixture; the mixture is formed into microparticles that are dispersed within bioabsorbable hydrogels, so as to release the water soluble therapeutic agents in a controlled fashion (see, e.g., 6,632,457). The in situ forming material may be a multi-component hydrogel system (see, e.g., U.S. Patent No. 6,379, 373). The in situ forming material may be a multi-arm block copolymer that includes a central core molecule, such as a residue of a polyol, and at least three copolymer arms covalently attached to the central core molecule, each copolymer arm comprising an inner hydrophobic polymer segment covalently attached to the central core molecule and an outer hydrophilic polymer segment covalently attached to the hydrophobic polymer segment, wherein the central core molecule and the hydrophobic polymer segment define a hydrophobic core region (see, e.g., 6,730,334). The in situ forming material may include a gel- forming macromer that includes at least four polymeric blocks, at least two of which are hydrophobic and at least one of which is hydrophilic, and including a crosslinkable group (see, e.g., 6,639,014). The in situ forming material may be a water-soluble macromer that includes at least one hydrolysable linkage formed from carbonate or dioxanone groups, at least one water-soluble polymeric block, and at least one polymerizable group (see, e.g., U.S. Patent No. 6,177,095). The in situ forming material may comprise polyoxyalkylene block copolymers that form weak physical crosslinks to provide gels having a paste-like consistency at physiological temperatures, (see, e.g., U.S. Patent No. 4,911,926). The in situ forming material may be a thermo-irreversible gel made from polyoxyalkylene polymers and ionic polysaccharides (see, e.g., U.S. Patent No. 5,126,141). The in situ forming material may be a gel forming composition that includes chitin derivatives (see, e.g., U.S. Patent No. 5,093,319), chitosan-coagulum (see, e.g., U.S. Patent No. 4,532,134), or hyaluronic acid (see, e.g., U.S. Patent No. 4,141 ,973). The in situ forming material may be an in situ modification of alginate (see, e.g., U.S. Patent No. 5,266,326 ). The in situ forming material may be formed from ethylenically unsaturated water soluble macromers that can be crosslinked in contact with tissues, cells, and bioactive molecules to form gels (see, e.g., U.S. Patent No. 5,573,934). The in situ forming material may include urethane prepolymers used in combination with an unsaturated cyano compound containing a cyano group attached to a carbon atom, such as cyano(meth)acrylic acids and esters thereof (see, e.g., U.S. Patent No. 4,740,534). The in situ forming material may be a biodegradable hydrogel that polymerizes by a photoinitiated free radical polymerization from water soluble macromers (see, e.g., U.S. Patent No. 5,410,016). The in situ forming material may be formed from a two component mixture including a first part comprising a serum albumin protein in an aqueous buffer having a pH in a range of about 8.0-11.0, and a second part comprising a water-compatible or water-soluble bifunctional crosslinking agent, (see, e.g., U.S. Patent No. 5,583,114).
In another aspect, in situ forming materials that can be used include those based on the crosslinking of proteins. For example, the in situ forming material may be a biodegradable hydrogel composed of a recombinant or natural human serum albumin and poly(ethylene) glycol polymer solution whereby upon mixing the solution cross-links to form a mechanical non-liquid covering structure which acts as a sealant. See, e.g., U.S. Patent No. 6,458,147 and 6,371 ,975. The in situ forming material may be composed of two separate mixtures based on fibrinogen and thrombin which are dispensed together to form a biological adhesive when intermixed either prior to or on the application site to form a fibrin sealant. See, e.g., U.S. Patent No. 6,764,467. The in situ forming material may be composed of ultrasonically treated collagen and albumin which form a viscous material that develops adhesive properties when crosslinked chemically with glutaraldehyde and amino acids or peptides. See, e.g., U.S. Patent No. 6,310,036. The in situ forming material may be a hydrated adhesive gel composed of an aqueous solution consisting essentially of a protein having amino groups at the side chains (e.g., gelatin, albumin) which is crosslinked with an N-hyd roxyim id oester compound. See, e.g., U.S. Patent No. 4,839,345. The in situ forming material may be a hydrogel prepared from a protein or polysaccharide backbone (e.g., albumin or polymannuronic acid) bonded to a cross-linking agent (e.g., polyvalent derivatives of polyethylene or polyalkylene glycol). See, e.g., U.S. Patent No. 5,514,379. The in situ forming material may be composed of a polymerizable collagen composition that is applied to the tissue and then exposed to an initiator to polymerize the collagen to form a seal over a wound opening in the tissue. See, e.g., U.S. Patent No. 5,874,537. The in situ forming material may be a two component mixture composed of a protein (e.g., serum albumin) in an aqueous buffer having a pH in the range of about 8.0-11.0 and a water-soluble bifunctional polyethylene oxide type crosslinking agent, which transforms from a liquid to a strong, flexible bonding composition to seal tissue in situ. See, e.g., U.S. Patents 5,583,114 and RE38158 and PCT Publication No. WO 96/03159. The in situ forming material may be composed of a protein, a surfactant, and a lipid in a liquid carrier, which is crosslinked by adding a crosslinker and used as a sealant or bonding agent in situ. See, e.g., U.S. Patent Application No. 2004/0063613A1 and PCT Publication Nos. WO 01/45761 and WO 03/090683. The in situ forming material may be composed of two enzyme-free liquid components that are mixed by dispensing the components into a catheter tube deployed at the vascular puncture site, wherein, upon mixing, the two liquid components chemically cross-link to form a mechanical non-liquid matrix that seals a vascular puncture site. See, e.g., U.S. Patent Application Nos. 2002/0161399A1 and 2001/0018598A1. The in situ forming material may be a cross-linked albumin composition composed of an albumin preparation and a carbodiimide preparation which are mixed under conditions that permit crosslinking of the albumin for use as a bioadhesive or sealant. See, e.g., PCT Publication No. WO 99/66964. The in situ forming material may be composed of collagen and a peroxidase and hydrogen peroxide, such that the collagen is crosslinked to from a semi-solid gel that seals a wound. See, e.g., PCT Publication No. WO 01/35882.
In another aspect, in situ forming materials that can be used include those based on isocyanate or isothiocyanate capped polymers. For example, the in situ forming material may be composed of isocyanate-capped polymers that are liquid compositions which form into a solid adhesive coating by in situ polymerization and crosslinking upon contact with body fluid or tissue. See, e.g., PCT Publication No. WO 04/021983. The in situ forming material may be a moisture-curing sealant composition composed of an active isocyanato-terminated isocyanate prepolymer containing a polyol component with a molecular weight of 2,000 to 20,000 and an isocyanurating catalyst agent. See, e.g., U.S. Patent No. 5,206,331.
In another embodiment, the reagents can undergo an electrophilic-nucleophilic reaction to produce a crosslinked matrix. Polymers containing and/or terminated with nucleophilic groups such as amine, sulfhydryl, hydroxyl, -PH2 or CO-NH-NH2 can be used as the nucleophilic reagents and polymers containing and/or terminated with electrophilic groups such as succinimidyl, carboxylic acid, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis can be used as the electrophilic reagents. For example, a 4- armed thiol derivatized poly(ethylene glycol) (e.g., pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl) can be reacted with a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra- succinimidyl glutarate) under basic conditions (pH > about 8). Representative examples of compositions that undergo such electrophilic-nucleophilic crosslinking reactions are described, for example, in U.S. Patent. Nos. 5,752,974; 5,807,581 ; 5,874,500; 5,936,035; 6,051 ,648; 6,165,489; 6,312,725; 6,458,889; 6,495,127; 6,534,591 ; 6,624,245; 6,566,406; 6,610,033; 6,632,457; and PCT Application Publication Nos. WO 04/060405 and WO 04/060346. Other examples of in situ forming materials that can be used include those based on the crosslinking of proteins (described in U.S. Patent Nos. RE38158; 4,839,345; 5,514,379, 5,583,114; 6,458,147; 6,371 ,975; U.S. Patent Application Publication Nos. 2002/0161399; 2001/0018598 and PCT Publication Nos. WO 03/090683; WO 01/45761 ; WO 99/66964 and WO 96/03159).
In another embodiment, the electrophilic- or nucleophilic- terminated polymers can further comprise a polymer that can enhance the mechanical and/or adhesive properties of the in situ forming compositions. This polymer can be a degradable or non-degradable polymer. For example, the polymer may be collagen or a collagen derivative, for example methylated collagen. An example of an in situ forming composition uses pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl) (4-armed thiol PEG), pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) (4-armed NHS PEG) and methylated collagen as the reactive reagents. This composition, when mixed with the appropriate buffers can produce a crosslinked hydrogel. (See, e.g., U.S. Patent Nos. 5,874,500; 6,051 ,648; 6,166,130; 5,565,519 and 6,312,725J. In another embodiment, the reagents that can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In the preferred embodiment, the 4 armed NHS-derivatized polyethylene glycol is applied to the tissue under basic conditions (pH > about 8). Other representative examples of compositions of this nature that may be used are disclosed in PCT Application Publication No. WO 04/060405 and WO 04/060346, and U.S. Patent Application No. 10/749,123.
In another embodiment, the in situ forming material polymer can be a polyester. Polyesters that can be used in in situ forming compositions include poly(hydroxyesters). In another embodiment, the polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, y-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one. Representative examples of these types of compositions are described in U.S.
Patent. Nos. 5,874,500; 5,936,035; 6,312,725; 6,495,127 and PCT Publication
Nos. WO 2004/028547.
In another embodiment, the electrophilic-terminated polymer can be partially or completely replaced by a small molecule or oligomer that comprises an electrophilic group (e.g., disuccinimidyl glutarate).
In another embodiment, the nucleophilic-terminated polymer can be partially or completely replaced by a small molecule or oligomer that comprises a nucleophilic group (e.g., dicysteine, dilysine, trilysine, etc.).
Other examples of in situ forming materials that can be used include those based on the crosslinking of proteins (described in, for example,
U.S. Patent Nos. RE38158; 4,839,345; 5,514,379, 5,583,114; 6,310,036;
6,458,147; 6,371 ,975; US Patent Application Publication Nos.
2004/0063613A1 , 2002/0161399A1 , and 2001/0018598A1 , and PCT
Publication Nos. WO 03/090683, WO 01/45761 , WO 99/66964, and WO 96/03159) and those based on isocyanate or isothiocyanate capped polymers
(see, e.g., PCT Publication No. WO 04/021983).
Other examples of in situ forming materials can include reagents that comprise one or more cyanoacrylate groups. These reagents can be used to prepare a poly(alkylcyanoacrylate) or poly(carboxyalkylcyanoacrylate) (e.g., poly(ethylcyanoacrylate), poly(butylcyanoacrylate), poly(isobutylcyanoacrylate), poly(hexylcyanoacrylate), poly(methoxypropylcyanoacrylate), and poly(octylcyanoacrylate)).
Examples of commercially available cyanoacrylates that can be used in the present invention include DERMABOND1 INDERMIL, GLUSTITCH, VETBOND, HISTOACRYL, TISSUMEND, HISTOACRYL BLUE and ORABASE
SOOTHE-N-SEAL LIQUID PROTECTANT. In another embodiment, the cyanoacrylate compositions may further comprise additives to stabilize the reagents and/or alter the rate of reaction of the cyanoacrylate, and/or plasticize the poly(cyanoacrylate), and/or alter the rate of degradation of the poly(cyanoacrylate). For example, a trimethylene carbonate based polymer or an oxalate polymer of poly(ethylene glycol) or a ε-caprolactone based copolymer may be mixed with a 2- alkoxyalkylcyanoacrylate (e.g., 2-methoxypropylcyanoacrylate). Representative examples of these compositions are described in U.S. Patent Nos. 5,350,798 and 6,299,631. In another embodiment, the cyanoacrylate composition can be prepared by capping heterochain polymers with a cyanoacrylate group. The cyanoacrylate-capped heterochain polymer preferably has at least two cyanoacrylate ester groups per chain. The heterochain polymer can comprise an absorbable poly(ester), poly(ester-carbonate), poly(ether-carbonate) and poly(ether-ester). The poly(ether-ester)s described in U.S. Patent Nos. 5,653,992 and 5,714,159 can also be used as the heterochain polymers. A triaxial poly(ε-caprolactone-co-trimethylene carbonate) is an example of a poly(ester-carbonate) that can be used. The heterochain polymer may be a polyether. Examples of polyethers that can be used include poly(ethylene glycol), poly(propylene glycol) and block copolymers of poly(ethylene glycol) and poly(propylene glycol) (e.g., PLURONICS group of polymers including but not limited to PLURONIC F127 or F68). Representative examples of these compositions are described in U.S. Patent No. 6,699,940.
Within another aspect of the invention, the biologically active ant- infective and/or fibrosis-inhibiting agent can be delivered with a non-polymeric compound (e.g., a carrier). These non-polymeric carriers can include sucrose derivatives (e.g., sucrose acetate isobutyrate, sucrose oleate), sterols such as cholesterol, stigmasterol, β-sitosterol, and estradiol; cholesteryl esters such as cholesteryl stearate; C12 -C24 fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, and lignoceric acid; Ci8 -C36 mono-, di- and triacylglycerides such as glyceryl monooleate, glyceryl monolinoleate, glyceryl monolaurate, glyceryl monodocosanoate, glyceryl monomyristate, glyceryl monodicenoate, glyceryl dipalmitate, glyceryl didocosanoate, glyceryl dimyristate, glyceryl didecenoate, glyceryl tridocosanoate, glyceryl trimyristate, glyceryl tridecenoate, glycerol tristearate and mixtures thereof; sucrose fatty acid esters such as sucrose distearate and sucrose palmitate; sorbitan fatty acid esters such as sorbitan monostearate, sorbitan monopalmitate and sorbitan tristearate; Ci6 -C18 fatty alcohols such as cetyl alcohol, myristyl alcohol, stearyl alcohol, and cetostearyl alcohol; esters of fatty alcohols and fatty acids such as cetyl palmitate and cetearyl palmitate; anhydrides of fatty acids such as stearic anhydride; phospholipids including phosphatidylcholine (lecithin), phosphatidylserine, phosphatidylethanolamine, phosphatidylinositol, and lysoderivatives thereof; sphingosine and derivatives thereof; spingomyelins such as stearyl, palmitoyl, and tricosanyl spingomyelins; ceramides such as stearyl and palmitoyl ceramides; glycosphingolipids; lanolin and lanolin alcohols, calcium phosphate, sintered and unscintered hydoxyapatite, zeolites; and combinations and mixtures thereof.
Representative examples of patents relating to non-polymeric delivery systems and the preparation include U.S. Patent Nos. 5,736,152; 5,888,533; 6,120,789; 5,968,542; and 5,747,058. Within certain embodiments of the invention, the therapeutic compositions are provided that include (i) a fibrosis-inhibiting agent and/or (ii) an anti-infective agent. The therapeutic compositions may include one or more additional therapeutic agents (such as described above), for example, antiinflammatory agents, anti-thrombotic agents, and/ or anti-platelet agents. Other agents that may be combined with the therapeutic compositions include, e.g., additional ingredients such as surfactants (e.g., PLURONICS, such as F-127, L-122, L-101 , L-92, L-81 , and L-61), preservatives, anti-oxidants.
In one aspect, the present invention provides compositions comprising i) an anti-fibrotic agent and ii) a polymer or a compound that forms a polymer in situ. The following are some, but by no means all, of the preferred anti-fibrotic agents and classes of anti-fibrotic agents that may be included in the inventive compositions:
1a) an anti-fibrotic agent that inhibits cell regeneration,
2a) an anti-fibrotic agent that inhibits angiogenesis, 3a) an anti-fibrotic agent that inhibits fibroblast migration,
4a) an anti-fibrotic agent that inhibits fibroblast proliferation,
5a) an anti-fibrotic agent that inhibits deposition of extracellular matrix,
6a) an anti-fibrotic agent inhibits tissue remodeling, 7a) an adensosine A2A receptor antagonist,
8a) an AKT inhibitor,
9a) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA),
10a) an alpha 4 integrin antagonist, 11 a) an alpha 7 nicotinic receptor agonist,
12a) an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE- 26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI- 8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567
(Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant
Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-
23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof,
13a) an apoptosis antagonist, 14a) an apoptosis activator,
15a) a beta 1 integrin antagonist,
16a) a beta tubulin inhibitor,
17a) a blocker of enzyme production in Hepatitis C,
18a) a Bruton's tyrosine kinase inhibitor, 19a) a calcineurin inhibitor,
20a) a caspase 3 inhibitor,
21 a) a CC chemokine receptor antagonist,
22a) a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, homoharringtonine, and an analogue or derivative thereof,
23a) a cathepsin B inhibitor,
24a) a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof, 25a) a cathepsin L inhibitor,
26a) a CD40 antagonist, 27a) a chemokine receptor agonist,
28a) a chymase inhibitor,
29a) a collagenase antagonist,
30a) a CXCR antagonist, 31 a) a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB- 286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof,
32a) a cyclooxygenase 1 inhibitor,
33a) a DHFR inhibitor,
34a) a dual integrin inhibitor,
35a) an elastase inhibitor, 36a) an elongation factor-1 alpha inhibitor,
37a) an endothelial growth factor antagonist,
38a) an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No. 125697-92-9), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof,
39a) an endotoxin antagonist,
40a) an epothilone and tubulin binder, 41a) an estrogen receptor antagonist,
42a) an FGF inhibitor,
43a) a farnexyl transferase inhibitor,
44a) a famesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B- 1055 (Yissum), and an analogue or derivative thereof,
45a) an FLT-3 kinase inhibitor,
46a) an FGF receptor kinase inhibitor, 47a) a fibrinogen antagonist selected from the group consisting of
AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, and an analogue or derivative thereof, 48a) a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro- 1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No. 12772-57-5), and an analogue or derivative thereof,
49a) a histone deacetylase inhibitor,
50a) an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No. 143201-11-0), and an analogue or derivative thereof, 51a) an ICAM inhibitor,
52a) an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof, 53a) an IL-2 inhibitor,
54a) an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA- 131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88
(Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof,
55a) an IMPDH (inosine monophosphate),
56a) an integrin antagonist, 57a) an interleukin antagonist,
58a) an inhibitor of type III receptor tyrosine kinase,
59a) an irreversible inhibitor of enzyme methionine aminopeptidase type 2,
60a) an isozyme selective delta protein kinase C inhibitor, 61a) a JAK3 enzyme inhibitor,
62a) a JNK inhibitor, 63a) a kinase inhibitor,
64a) a kinesin antagonist,
65a) a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotrϊene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912- 92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3
(Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof,
66a) a MAP kinase inhibitor,
67a) a matrix metalloproteinase inhibitor, 68a) an MCP-CCR2 inhibitor,
69a) an mTOR inhibitor,
70a) an mTOR kinase inhibitor,
71a) a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, dolastatin 15 (CAS No. 123884-00-4), vincamine, and an analogue or derivative thereof, 72a) an MIF inhibitor, 73a) an MMP inhibitor, 74a) a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof,
75a) an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), Bay 11-7085, and an analogue or derivative thereof, 76a) a nitric oxide agonist, 77a) an ornithine decarboxylase inhibitor, 78a) a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS- 2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74-6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof, 79a) a palmitoyl-protein thioesterase inhibitor, 80a) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof, 81a) a peroxisome proliferators-activated receptor agonist selected from the group consisting of (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70- 4 and 112529-15-4), ACTOPLUS M ET from And rx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0)
(GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY- 14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof,
82a) a phosphatase inhibitor,
83a) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66- 5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof, 84a) a PKC inhibitor,
85a) a platelet activating factor antagonist,
86a) a platelet-derived growth factor receptor kinase inhibitor,
87a) a prolyl hydroxylase inhibitor,
88a) a polymorphonuclear neutrophil inhibitor, 89a) a protein kinase B inhibitor,
90a) a protein kinase C stimulant, 91a) a purine nucleoside analogue,
92a) a purinoreceptor P2X antagonist,
93a) a Raf kinase inhibitor,
94a) a reversible inhibitor of ErbB1 and ErbB2, 95a) a ribonucleoside triphosphate reductase inhibitor,
96a) an SDF-1 antagonist,
97a) a sheddase inhibitor,
98a) an SRC inhibitor,
99a) a stromelysin inhibitor, 100a) an Syk kinase inhibitor,
101a) a telomerase inhibitor,
102a) a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof,
103a) a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti- TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006
(Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS)1 infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11 -3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Ys Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof, 104a) a tumor necrosis factor antagonist,
105a) a Toll receptor inhibitor, 106a) a tubulin antagonist,
107a) a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS- 354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL- 993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP- 5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR- 200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP- 724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax- EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalaiide F (CAS No. 149204- 42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50- 1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL- 647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), herbimycin A, and an analogue or derivative thereof,
108a) a VEGF inhibitor, 109a) a vitamin D receptor agonist, 110a) ZD-6474 (an angiogenesis inhibitor), 111a) AP-23573 (an mTOR inhibitor),
112a) synthadotin (a tubulin antagonist), 113a) S-0885 (a collagenase inhibitor), 114a) aplidine (an elongation factor-1 alpha inhibitor), 115a) ixabepilone (an epithilone), 116a) IDN-5390 (an angiogenesis inhibitor and an FGF inhibitor),
117a) SB-2723005 (an angiogenesis inhibitor), 118a) ABT-518 (an angiogenesis inhibitor), 119a) combretastatin (an angiogenesis inhibitor), 120a) anecortave acetate (an angiogenesis inhibitor), 121a) SB-715992 (a kinesin antagonist),
122a) temsirolimus (an mTOR inhibitor), 123a) adalimumab (a TNFα antagonist), 124a) erucylphosphocholine (an ATK inhibitor), 125a) alphastatin (an angiogenesis inhibitor), 126a) bortezomib (an NF Kappa B inhibitor),
127a) etanercept (a TNFα antagonist and TACE inhibitor), 128a) humicade (a TNFα inhibitor), 129a) gefitinib (a tyrosine kinase inhibitor), 130a) a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones),
131a) an alpha adrenergic receptor antagonist,
132a) an anti-psychotic compound, 133a) a CaM kinase Il inhibitor,
134a) a G protein agonist,
135a) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof, 136a) an anti-microbial agent,
137a) a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770- 78-7), aurintricarboxylic acid, and an analogue or derivative thereof,
138a) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof,
139a) a D2 dopamine receptor antagonist,
140a) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor,
141a) a dopamine antagonist, an anesthetic compound, 142a) a clotting factor,
143a) a lysyl hydrolase inhibitor, 144a) a muscarinic receptor inhibitor,
145a) a superoxide anion generator,
146a) a steroid,
147a) an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof, 148a) a diuretic,
149a) an anti-coagulant,
150a) a cyclic GMP agonist,
151a) an adenylate cyclase agonist,
152a) an antioxidant, 153a) a nitric oxide synthase inhibitor,
154a) an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof,
155a) a DNA synthesis inhibitor, 156a) a DNA alkylating agent selected from dacarbazine (CAS
No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof,
157a) a DNA methylation inhibitor,
158a) a NSAID agent, 159a) a peptidylglycine alpha-hydroxylating monooxygenase inhibitor,
160a) an MEK1/MEK 2 inhibitor,
161a) a NO synthase inhibitor,
162a) a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof,
163a) an ACE inhibitor, 164a) a glycosyϊation inhibitor,
165a) an intracellular calcium influx inhibitor,
166a) an anti-emetic agent,
167a) an acetylcholinesterase inhibitor, 168a) an ALK-5 receptor antagonist,
169a) a RAR/RXT antagonist,
170a) an elF-2a inhibitor,
171a) an S-adenosyl-L-homocysteine hydrolase inhibitor,
172a) an estrogen agonist, 173a) a serotonin receptor inhibitor,
174a) an anti-thrombotic agent,
175a) a tryptase inhibitor,
176a) a pesticide,
177a) a bone mineralization promoter, 178a) a bisphosphonate compound selected from risedronate and r derivative thereof,
179a) an anti-inflammatory compound,
180a) a DNA methylation promoter,
181a) an anti-spasmodic agent, 182a) a protein synthesis inhibitor,
183a) an α-glucosidase inhibitor,
184a) a calcium channel blocker,
185a) a pyruvate dehydrogenase activator,
186a) a prostaglandin inhibitor, 187a) a sodium channel inhibitor,
188a) a serine protease inhibitor,
189a) an intracellular calcium flux inhibitor,
190a) a JAK2 inhibitor;
191a) an androgen inhibitor, 192a) an aromatase inhibitor,
193a) an anti-viral agent, 194a) a 5-HT inhibitor,
195a) an FXR antagonist,
196a) an actin polymerization and stabilization promoter,
197a) an AXOR12 agonist, 198a) an angiotensin Il receptor agonist,
199a) a platelet aggregation inhibitor,
200a) a CB1/CB2 receptor agonist,
201a) a norepinephrine reuptake inhibitor,
202a) a selective serotonin reuptake inhibitor, 203a) a reducing agent,
204a) isotretinoin,
205a) radicicol,
206a) clobetasol propionate,
207a) homoharringtonine, 208a) trichostatin A,
209a) brefeldin A,
210a) thapsigargin,
211a) dolastatin 15,
212a) cerivastatin, 213a) jasplakinolide,
214a) herbimycin A,
215a) pirfenidone,
216a) vinorelbine,
217a) 17-DMAG, 218a) tacrolimus,
219a) loteprednol etabonate,
220a) juglone,
221a) prednisolone,
222a) puromycin, 223a) 3-BAABE,
224a) cladribine, 225a) mannose-6-phosphate,
226a) 5-azacytidine,
227a) Ly333531 (ruboxistaurin),
228a) simvastatin, and 229a) an immuno-modulator selected from Bay 11 -7085, (-)- arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
As mentioned above, the present invention provides compositions comprising each of the foregoing 229 (i.e., 1a through 229a) listed anti-fibrotic agents or classes of anti-fibrotic agents, with each of the following 97 (i.e., 1b through 97b) polymers and compounds:
1b. A crosslinked polymer.
2b. A polymer that reacts with mammalian tissue.
3b. A polymer that is a naturally occurring polymer.
4b. A polymer that is a protein. 5b. A polymer that is a carbohydrate.
6b. A polymer that is biodegradable.
7b. A polymer that is crosslinked and biodegradable.
8b. A polymer that nonbiodegradable.
9b. Collagen. 10b. Methylated collagen.
11b. Fibrinogen.
12b. Thrombin.
13b. Albumin.
14b. Plasminogen. 15b. von Willebrands factor.
16b. Factor VIII.
17b. Hypoallergenic collagen.
18b. Atelopeptidic collagen.
19b. Telopeptide collagen. 20b. Crosslinked collagen.
21b. Aprotinin. 22b. Gelatin.
23b. A protein conjugate.
24b. A gelatin conjugate.
25b. Hyaluronic acid. 26b. A hyaluronic acid derivative.
27b. A synthetic polymer.
28b. A polymer formed from reactants comprising a synthetic isocyanate-containing compound.
29b. A synthetic isocyanate-containing compound. 30b. A polymer formed from reactants comprising a synthetic thiol-containing compound.
31 b. A synthetic thiol-containing compound.
32b. A polymer formed from reactants comprising a synthetic compound containing at least two thiol groups. 33b. A synthetic compound containing at least two thiol groups.
34b. A polymer formed from reactants comprising a synthetic compound containing at least three thiol groups.
35b. A synthetic compound containing at least three thiol groups. 36b. A polymer formed from reactants comprising a synthetic compound containing at least four thiol groups.
37b. A synthetic compound containing at least four thiol groups.
38b. A polymer formed from reactants comprising a synthetic amino-containing compound. 39b. A synthetic amino-containing compound.
40b. A polymer formed from reactants comprising a synthetic compound containing at least two amino groups.
41 b. A synthetic compound containing at least two amino groups. 42b. A polymer formed from reactants comprising a synthetic compound containing at least three amino groups. 43b. A synthetic compound containing at least three amino groups..
44b. A polymer formed from reactants comprising a synthetic compound containing at least four amino groups. 45b. A synthetic compound containing at least four amino groups.
46b. A polymer formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
47b. A synthetic compound comprising a carbonyl-oxygen- succinimidyl group.
48b. A polymer formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
49b. A synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups. 50b. A polymer formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
51 b. A synthetic compound comprising at least three carbonyl- oxygen-succinimidyl groups.
52b. A polymer formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
53b. A synthetic compound comprising at least four carbonyl- oxygen-succinimidyl groups.
54b. A polymer formed from from reactants comprising a synthetic polyalkylene oxide-containing compound. 55b. A synthetic polyalkylene oxide-containing compound.
56b. A polymer formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
57b. A synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks. 58b. A polymer formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
59b. A synthetic polyalkylene oxide-containing compound having reactive amino groups. 60b. A polymer formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
61b. A synthetic polyalkylene oxide-containing compound having reactive thiol groups.
62b. A polymer formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen- succinimidyl groups.
63b. A synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
64b. A polymer formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
65b. A synthetic compound comprising a biodegradable polyester block.
66b. A polymer formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide. 67b. A synthetic polymer formed in whole or part from lactic acid or lactide.
68b. A polymer formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
69b. A synthetic polymer formed in whole or part from glycolic acid or glycolide.
70b. A polymer formed from reactants comprising polylysine.
71b. Polylysine.
72b. A polymer formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion. 73b. A polymer formed from reactants comprising (a) protein and (b) polylysine. 74b. A polymer formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
75b. A polymer formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 76b. A polymer formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
77b. A polymer formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon- caprolactone.
78b. A polymer formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
79b. A polymer formed from reactants comprising (a) collagen and (b) polylysine. 80b. A polymer formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
81b. A polymer formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
82b. A polymer formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
83b. A polymer formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon- caprolactone. 84b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
85b. A polymer formed from reactants comprising (a) methylated collagen and (b) polylysine. 86b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 87b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
88b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen- succinimide groups.
89b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone. 90b. A polymer formed from reactants comprising hyaluronic acid.
91b. A polymer formed from reactants comprising a hyaluronic acid derivative.
92b. A polymer formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
93b. Pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
94b. A polymer formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
95b. Pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
96b. A polymer formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups. 97b. A mixture of (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecularweight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
As mentioned above, the present invention provides compositions comprising each ofthe foregoing 229 (1a through 229a) listed anti-fibrotic agents or classes ofanti-fibrotic agents, with each oftheforegoing 97 (1b through 97b) polymers and compounds: Thus, in separate aspects, the invention provides 229 times 97 = 22,213 described compositions. In otherwords, each ofthe following is a distinct aspect ofthe present invention: 1a+1b, 2a+1b, 3a+1b, 4a+1b, 5a+1b, 6a+1b, 7a+1b, 8a+1b, 9a+1b, 10a+1b, 11a+1b, 12a+1b, 13a+1b, 14a+1b, 15a+1b, 16a+1b, 17a+1b, 18a+1b, 19a+1b, 20a+1b, 21a+1b, 22a+1b, 23a+1b, 24a+1b, 25a+1b, 26a+1b, 27a+1b, 28a+1b, 29a+1b, 30a+1b, 31a+1b, 32a+1b, 33a+1b, 34a+1b, 35a+1b, 36a+1b, 37a+1b, 38a+1b, 39a+1b, 40a+1b, 41a+1b, 42a+1b, 43a+1b, 44a+1b, 45a+1b, 46a+1b, 47a+1b, 48a+1b, 49a+1b, 50a+1b, 51a+1b, 52a+1b, 53a+1b, 54a+1b, 55a+1b, 56a+1b, 57a+1b, 58a+1b, 59a+1b, 60a+1b, 61a+1b, 62a+1b, 63a+1b, 64a+1b, 65a+1b, 66a+1b, 67a+1b, 68a+1b, 69a+1b, 70a+1b, 71a+1b, 72a+1b, 73a+1b, 74a+1b, 75a+1b, 76a+1b, 77a+1b, 78a+1b, 79a+1b, 80a+1b, 81a+1b, 82a+1b, 83a+1b, 84a+1b, 85a+1b, 86a+1b, 87a+1b, 88a+1b, 89a+1b, 90a+1b, 91a+1b, 92a+1b, 93a+1b, 94a+1b, 95a+1b, 96a+1b, 97a+1b, 98a+1b, 99a+1b, 100a+1b, 101a+1b, 102a+1b, 103a+1b, 104a+1b, 105a+1b, 106a+1b, 107a+1b, 108a+1b, 109a+1b, 110a+1b, 111a+1b, 112a+1b, 113a+1b, 114a+1b, 115a+1b, 116a+1b, 117a+1b, 118a+1b, 119a+1b, 120a+1b, 121a+1b, 122a+1b, 123a+1b, 124a+1b, 125a+1b, 126a+1b, 127a+1b, 128a+1b, 129a+1b, 130a+1b, 131a+1b, 132a+1b, 133a+1b, 134a+1b, 135a+1b, 136a+1b, 137a+1b, 138a+1b, 139a+1b, 140a+1b, 141a+1b, 142a+1b, 143a+1b, 144a+1b, 145a+1b, 146a+1b, 147a+1b, 148a+1b, 149a+1b, 150a+1b, 151a+1b, 152a+1b, 153a+1b, 154a+1b, 155a+1b, 156a+1b, 157a+1b, 158a+1b, 159a+1b, 160a+1b, 161a+1b, 162a+1b, 163a+1b, 164a+1b, 165a+1b, 166a+1b, 167a+1b, 168a+1b, 169a+1b, 170a+1b, 171a+1b, 172a+1b, 173a+1b, 174a+1b, 175a+1b, 176a+1b, 177a+1b, 178a+1b, 179a+1b, 180a+1b, 181a+1b, 182a+1b, 183a+1b, 184a+1b, 185a+1b, 186a+1b, 187a+1b, 188a+1b, 189a+1b, 190a+1b, 191a+1b, 192a+1b, 193a+1b, 194a+1b, 95a+1b, 96a+1b, 97a+1b, 98a+1b, 99a+1b, 200a+1b, 201a+1b, 202a+1b, 203a+1b, 204a+1b, 205a+1b, 206a+1b, 207a+1b, 208a+1b, 209a+1b, 210a+1b, 211a+1b, 212a+1b, 213a+1b, 214a+1b( 215a+1b, 216a+1b, 217a+1b, 218a+1b, 219a+1b, 220a+1b, 221a+1b, 222a+1b, 223a+1b, 224a+1b, 225a+1b, 226a+1b, 227a+1b, 228a+1b, 229a+1b, eto.
All U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety.
The present invention, in various aspects, provides the following itemized embodiments.
1. A device, comprising a sensor and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2. The device of item 1 wherein the agent is an adensosine A2A receptor antagonist.
3. The device of item 1 wherein the agent is an AKT inhibitor.
4. The device of item 1 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 5. The device of item 1 wherein the agent is an alpha 4 integrin antagonist.
6. The device of item 1 wherein the agent is an alpha 7 nicotinic receptor agonist.
7. The device of item 1 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8. The device of item 1 wherein the agent is an apoptosis antagonist.
9. The device of item 1 wherein the agent is an apoptosis activator.
10. The device of item 1 wherein the agent is a beta 1 integrin antagonist.
11. The device of item 1 wherein the agent is a beta tubulin inhibitor.
12. The device of item 1 wherein the agent is a blocker of enzyme production in Hepatitis C.
13. The device of item 1 wherein the agent is a Bruton's tyrosine kinase inhibitor.
14. The device of item 1 wherein the agent is a calcineurin inhibitor.
15. The device of item 1 wherein the agent is a caspase 3 inhibitor.
16. The device of item 1 wherein the agent is a CC chemokine receptor antagonist.
17. The device of item 1 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 18. The device of item 1 wherein the agent is a cathepsin B inhibitor.
19. The device of item 1 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL- 022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
20. The device of item 1 wherein the agent is a cathepsin L inhibitor.
21. The device of item 1 wherein the agent is a CD40 antagonist.
22. The device of item 1 wherein the agent is a chemokine receptor agonist.
23. The device of item 1 wherein the agent is a chymase inhibitor.
24. The device of item 1 wherein the agent is a collagenase antagonist.
25. The device of item 1 wherein the agent is a CXCR antagonist.
26. The device of item 1 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
27. The device of item 1 wherein the agent is a cyclooxygenase 1 inhibitor.
28. The device of item 1 wherein the agent is a DHFR inhibitor.
29. The device of item 1 wherein the agent is a dual integrin inhibitor.
30. The device of item 1 wherein the agent is an elastase inhibitor.
31. The device of item 1 wherein the agent is an elongation factor-1 alpha inhibitor.
32. The device of item 1 wherein the agent is an endothelial growth factor antagonist.
33. The device of item 1 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57- 9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
34. The device of item 1 wherein the agent is an endotoxin antagonist. 35. The device of item 1 wherein the agent is an epothilone and tubulin binder.
36. The device of item 1 wherein the agent is an estrogen receptor antagonist.
37. The device of item 1 wherein the agent is an FGF inhibitor.
38. The device of item 1 wherein the agent is a farnexyl transferase inhibitor.
39. The device of item 1 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
40. The device of item 1 wherein the agent is an FLT-3 kinase inhibitor.
41. The device of item 1 wherein the agent is an FGF receptor kinase inhibitor.
42. The device of item 1 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-
13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
43. The device of item 1 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
44. The device of item 1 wherein the agent is a histone deacetylase inhibitor.
45. The device of item 1 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
46. The device of item 1 wherein the agent is an ICAM inhibitor.
47. The device of item 1 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
48. The device of item 1 wherein the agent is an IL-2 inhibitor.
49. The device of item 1 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
50. The device of item 1 wherein the agent is an IMPDH (inosine monophosphate).
51. The device of item 1 wherein the agent is an integrin antagonist.
52. The device of item 1 wherein the agent is an interleukin antagonist.
53. The device of item 1 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
54. The device of item 1 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
55. The device of item 1 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
56. The device of item 1 wherein the agent a JAK3 enzyme inhibitor.
57. The device of item 1 wherein the agent is a JNK inhibitor.
58. The device of item 1 wherein the agent is a kinase inhibitor.
59. The device of item 1 wherein the agent is kinesin antagonist. 60. The device of item 1 wherein the agent is a kinesin antagonist.
61. The device of item 1 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
62. The device of item 1 wherein the agent is an MAP kinase inhibitor.
63. The device of item 1 wherein the agent is a matrix metalloproteinase inhibitor.
64. The device of item 1 wherein the agent is an MCP-CCR2 inhibitor.
65. The device of item 1 wherein the agent is an mTOR inhibitor.
66. The device of item 1 wherein the agent is an mTOR kinase inhibitor. 67. The device of item 1 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
68. The device of item 1 wherein the agent is an MIF inhibitor.
69. The device of item 1 wherein the agent is an MMP inhibitor.
70. The device of item 1 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL- 105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
71. The device of item 1 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi- Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (lnflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104- 49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
72. The device of item 1 wherein the agent is a nitric oxide agonist.
73. The device of item 1 wherein the agent is an ornithine decarboxylase inhibitor.
74. The device of item 1 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
75. The device of item 1 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
76. The device of item 1 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
77. The device of item 1 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
78. The device of item 1 wherein the agent is a phosphatase inhibitor.
79. The device of item 1 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
80. The device of item 1 wherein the agent is a PKC inhibitor.
81. The device of item 1 wherein the agent is a platelet activating factor antagonist.
82. The device of item 1 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
83. The device of item 1 wherein the agent is a prolyl hydroxylase inhibitor.
84. The device of item 1 wherein the agent is a polymorphonuclear neutrophil inhibitor.
85. The device of item 1 wherein the agent is a protein kinase
B inhibitor.
86. The device of item 1 wherein the agent is a protein kinase C stimulant.
87. The device of item 1 wherein the agent is a purine nucleoside analogue. 88. The device of item 1 wherein the agent is a purinoreceptor P2X antagonist.
89. The device of item 1 wherein the agent is a Raf kinase inhibitor.
90. The device of item 1 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
91. The device of item 1 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
92. The device of item 1 wherein the agent is an SDF-1 antagonist.
93. The device of item 1 wherein the agent is a sheddase inhibitor.
94. The device of item 1 wherein the agent is an SRC inhibitor.
95. The device of item 1 wherein the agent is a stromelysin inhibitor.
96. The device of item 1 wherein the agent is an Syk kinase inhibitor.
97. The device of item 1 wherein the agent is a telomerase inhibitor.
98. The device of item 1 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
99. The device of item 1 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC)1 RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
100. The device of item 1 wherein the agent is a Toll receptor inhibitor.
101. The device of item 1 wherein the agent is a tubulin antagonist.
102. The device of item 1 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
103. The device of item 1 wherein the agent is a VEGF inhibitor.
104. The device of item 1 wherein the agent is a vitamin D receptor agonist. 105. The device of item 1 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
106. The device of item 1 wherein the agent is AP-23573 (an mTOR inhibitor).
107. The device of item 1 wherein the agent is synthadotin (a tubulin antagonist).
108. The device of item 1 wherein the agent is S-0885 (a collagenase inhibitor).
109. The device of item 1 wherein the agent is aplidine (an elongation factor- 1 alpha inhibitor).
110. The device of item 1 wherein the agent is ixabepilone (an epithilone).
111. The device of item 1 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
112. The device of item 1 wherein the agent is SB-2723005
(an angiogenesis inhibitor).
113. The device of item 1 wherein the agent is ABT-518 (an angiogenesis inhibitor).
114. The device of item 1 wherein the agent is combretastatin (an angiogenesis inhibitor).
115. The device of item 1 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
116. The device of item 1 wherein the agent is SB-715992 (a kinesin antagonist). 117. The device of item 1 wherein the agent is temsirolimus (an mTOR inhibitor).
118. The device of item 1 wherein the agent is adalimumab (a TNFα antagonist).
119. The device of item 1 , further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
120. The device of item 1, further comprising a coating, wherein the coating comprises the anti-scarring agent.
121. The device of item 1 , further comprising a coating, wherein the coating is disposed on a surface of the device.
122. The device of item 1 , further comprising a coating, wherein the coating directly contacts the device.
123. The device of item 1, further comprising a coating, wherein the coating indirectly contacts the device.
124. The device of item 1, further comprising a coating, wherein the coating partially covers the device.
125. The device of item 1 , further comprising a coating, wherein the coating completely covers the device.
126. The device of item 1 , further comprising a coating, wherein the coating is a uniform coating.
127. The device of item 1 , further comprising a coating, wherein the coating is a non-uniform coating.
128. The device of item 1, further comprising a coating, wherein the coating is a discontinuous coating. 129. The device of item 1 , further comprising a coating, wherein the coating is a patterned coating.
130. The device of item 1 , further comprising a coating, wherein the coating has a thickness of 100 μm or less.
131. The device of item 1 , further comprising a coating, wherein the coating has a thickness of 10 μm or less.
132. The device of item 1 , further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
133. The device of item 1 , further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
134. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
135. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
136. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
137. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
138. The device of item 1 , further comprising a coating, wherein the coating further comprises a polymer. 139. The device of item 1 , further comprising a first coating having a first composition and the second coating having a second composition.
140. The device of item 1 , further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
141. The device of item 1 , further comprising a polymer.
142. The device of item 1 , further comprising a polymeric carrier.
143. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
144. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
145. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
146. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
147. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
148. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
149. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer. 150. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
151. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
152. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
153. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
154. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
155. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
156. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
157. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
158. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
159. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
160. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 161. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
162. The device of item 1 , further comprising a lubricious coating.
163. The device of item 1 wherein the anti-scarring agent is located within pores or holes of the device.
164. The device of item 1 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
165. The device of item 1, further comprising a second pharmaceutically active agent.
166. The device of item 1 , further comprising an antiinflammatory agent.
167. The device of item 1 , further comprising an agent that inhibits infection.
168. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
169. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
170. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
171. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
172. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU). 173. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
174. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is methotrexate.
175. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
176. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is etoposide.
177. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
178. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
179. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
180. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is cisplatin.
181. The device of item 1 , further comprising an antithrombotic agent.
182. The device of item 1 , further comprising a visualization agent.
183. The device of item 1 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 184. The device of item 1 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
185. The device of item 1 , further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
186. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
187. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
188. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
189. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
190. The device of item 1 , further comprising an echogenic material.
191. The device of item 1 , further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
192. The device of item 1 wherein the device is sterile.
193. The device of item 1 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
194. The device of item 1 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device. 195. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
196. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
197. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
198. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
199. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
200. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
201. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
202. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
203. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate. 204. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
205. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
206. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
207. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
208. The device of item 1 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
209. The device of item 1 wherein the device comprises about
10 μg to about 10 mg of the anti-scarring agent.
210. The device of item 1 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
211. The device of item 1 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
212. The device of item 1 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
213. The device of item 1 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 214. The device of item 1 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
215. The device of item 1 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
216. The device of item 1 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
217. The device of item 1 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
218. The device of item 1 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
219. The device of item 1 wherein the agent or the composition is affixed to the sensor.
220. The device of item 1 wherein the agent or the composition is covalently attached to the sensor.
221. The device of item 1 wherein the agent or the composition is non-covalently attached to the sensor.
222. The device of item 1 further comprising a coating that absorbs the agent or the composition.
223. The device of item 1 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition. 224. The device of item 1 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
225. The device of item 1 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
226. The device of item 1 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
227. The device of item 1 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
228. The device of item 1 further comprising a pump that is linked to the sensor.
229. The device of any one of items 1-228 wherein the sensor is a blood or tissue glucose monitor.
230. The device of any one of items 1-228 wherein the sensor is an electrolyte sensor.
231. The device of any one of items 1-228 wherein the sensor is a blood constituent sensor.
232. The device of any one of items 1-228 wherein the sensor is a temperature sensor.
233. The device of any one of items 1-228 wherein the sensor is a pH sensor.
234. The device of any one of items 1-228 wherein the sensor is an optical sensor.
235. The device of any one of items 1-228 wherein the sensor is an amperometric sensor. 236. The device of any one of items 1-228 wherein the sensor is a pressure sensor.
237. The device of any one of items 1-228 wherein the sensor is a biosensor.
238. The device of any one of items 1-228 wherein the sensor is a sensing transponder.
239. The device of any one of items 1-228 wherein the sensor is a strain sensor.
240. The device of any one of items 1-228 wherein the sensor is a magnetoresistive sensor.
241. The device of any one of items 1-228 wherein the sensor is a cardiac sensor.
242. The device of any one of items 1-228 wherein the sensor is a respiratory sensor.
243. The device of any one of items 1-228 wherein the sensor is an auditory sensor.
244. The device of any one of items 1-228 wherein the sensor is a metabolite sensor.
245. The device of any one of items 1-228 wherein the sensor detects mechanical changes.
246. The device of any one of items 1-228 wherein the sensor detects physical changes.
247. The device of any one of items 1-228 wherein the sensor detects electrochemical changes. 248. The device of any one of items 1-228 wherein the sensor detects magnetic changes.
249. The device of any one of items 1-228 wherein the sensor detects acceleration changes.
250. The device of any one of items 1-228 wherein the sensor detects ionizing radiation changes.
251. The device of any one of items 1-228 wherein the sensor detects acoustic wave changes.
252. The device of any one of items 1-228 wherein the sensor detects chemical changes.
253. The device of any one of items 1-228 wherein the sensor detects drug concentration changes.
254. The device of any one of items 1-228 wherein the sensor detects hormone changes.
255. The device of any one of items 1-228 wherein the sensor detects barometric changes.
256. A device, comprising a blood or tissue glucose monitor (i.e., a sensor) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
257. The device of item 256 wherein the agent is an adensosine A2A receptor antagonist.
258. The device of item 256 wherein the agent is an AKT inhibitor. 259. The device of item 256 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
260. The device of item 256 wherein the agent is an alpha 4 integrin antagonist.
261. The device of item 256 wherein the agent is an alpha 7 nicotinic receptor agonist.
262. The device of item 256 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DACiantiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant
Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
263. The device of item 256 wherein the agent is an apoptosis antagonist.
264. The device of item 256 wherein the agent is an apoptosis activator.
265. The device of item 256 wherein the agent is a beta 1 integrin antagonist.
266. The device of item 256 wherein the agent is a beta tubulin inhibitor.
267. The device of item 256 wherein the agent is a blocker of enzyme production in Hepatitis C.
268. The device of item 256 wherein the agent is a Bruton's tyrosine kinase inhibitor.
269. The device of item 256 wherein the agent is a calcineurin inhibitor.
270. The device of item 256 wherein the agent is a caspase 3 inhibitor.
271. The device of item 256 wherein the agent is a CC chemokine receptor antagonist. 272. The device of item 256 wherein the agent is a ceil cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
273. The device of item 256 wherein the agent is a cathepsin B inhibitor.
274. The device of item 256 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
275. The device of item 256 wherein the agent is a cathepsin L inhibitor.
276. The device of item 256 wherein the agent is a CD40 antagonist.
277. The device of item 256 wherein the agent is a chemokine receptor agonist.
278. The device of item 256 wherein the agent is a chymase inhibitor.
279. The device of item 256 wherein the agent is a collagenase antagonist.
280. The device of item 256 wherein the agent is a CXCR antagonist.
281. The device of item 256 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (Eli Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
282. The device of item 256 wherein the agent is a cyclooxygenase 1 inhibitor.
283. The device of item 256 wherein the agent is a DHFR inhibitor.
284. The device of item 256 wherein the agent is a dual integrin inhibitor.
285. The device of item 256 wherein the agent is an elastase inhibitor.
286. The device of item 256 wherein the agent is an elongation factor- 1 alpha inhibitor.
287. The device of item 256 wherein the agent is an endothelial growth factor antagonist.
288. The device of item 256 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
289. The device of item 256 wherein the agent is an endotoxin antagonist.
290. The device of item 256 wherein the agent is an epothilone and tubulin binder.
291. The device of item 256 wherein the agent is an estrogen receptor antagonist.
292. The device of item 256 wherein the agent is an FGF inhibitor.
293. The device of item 256 wherein the agent is a farnexyl transferase inhibitor.
294. The device of item 256 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
295. The device of item 256 wherein the agent is an FLT-3 kinase inhibitor.
296. The device of item 256 wherein the agent is an FGF receptor kinase inhibitor.
297. The device of item 256 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
298. The device of item 256 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
299. The device of item 256 wherein the agent is a histone deacetylase inhibitor.
300. The device of item 256 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
301. The device of item 256 wherein the agent is an ICAM inhibitor.
302. The device of item 256 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
303. The device of item 256 wherein the agent is an IL-2 inhibitor.
304. The device of item 256 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
305. The device of item 256 wherein the agent is an IMPDH (inosine monophosphate).
306. The device of item 256 wherein the agent is an integrin antagonist.
307. The device of item 256 wherein the agent is an interleukin antagonist.
308. The device of item 256 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
309. The device of item 256 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
310. The device of item 256 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
311. The device of item 256 wherein the agent a JAK3 enzyme inhibitor. 312. The device of item 256 wherein the agent is a JNK inhibitor.
313. The device of item 256 wherein the agent is a kinase inhibitor.
314. The device of item 256 wherein the agent is kinesin antagonist.
315. The device of item 256 wherein the agent is a kinesin antagonist.
316. The device of item 256 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
317. The device of item 256 wherein the agent is an MAP kinase inhibitor.
318. The device of item 256 wherein the agent is a matrix metalloproteinase inhibitor. 319. The device of item 256 wherein the agent is an MCP- CCR2 inhibitor.
320. The device of item 256 wherein the agent is an mTOR inhibitor.
321. The device of item 256 wherein the agent is an mTOR kinase inhibitor.
322. The device of item 256 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR- 112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
323. The device of item 256 wherein the agent is an MIF inhibitor.
324. The device of item 256 wherein the agent is an MMP inhibitor.
325. The device of item 256 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-
105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
326. The device of item 256 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547
(Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE)1 IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG)1 NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
327. The device of item 256 wherein the agent is a nitric oxide agonist.
328. The device of item 256 wherein the agent is an ornithine decarboxylase inhibitor.
329. The device of item 256 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
330. The device of item 256 wherein the agent is a paimitoyl- protein thioesterase inhibitor.
331. The device of item 256 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
332. The device of item 256 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET
(GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
333. The device of item 256 wherein the agent is a phosphatase inhibitor. 334. The device of item 256 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
335. The device of item 256 wherein the agent is a PKC inhibitor.
336. The device of item 256 wherein the agent is a platelet activating factor antagonist.
337. The device of item 256 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
338. The device of item 256 wherein the agent is a prolyl hydroxylase inhibitor. 339. The device of item 256 wherein the agent is a polymorphonuclear neutrophil inhibitor.
340. The device of item 256 wherein the agent is a protein kinase B inhibitor.
341. The device of item 256 wherein the agent is a protein kinase C stimulant.
342. The device of item 256 wherein the agent is a purine nucleoside analogue.
343. The device of item 256 wherein the agent is a purinoreceptor P2X antagonist.
344. The device of item 256 wherein the agent is a Raf kinase inhibitor.
345. The device of item 256 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
346. The device of item 256 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
347. The device of item 256 wherein the agent is an SDF-1 antagonist.
348. The device of item 256 wherein the agent is a sheddase inhibitor.
349. The device of item 256 wherein the agent is an SRC inhibitor.
350. The device of item 256 wherein the agent is a stromelysin inhibitor. 351. The device of item 256 wherein the agent is an Syk kinase inhibitor.
352. The device of item 256 wherein the agent is a telomerase inhibitor.
353. The device of item 256 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
354. The device of item 256 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
355. The device of item 256 wherein the agent is a Toll receptor inhibitor.
356. The device of item 256 wherein the agent is a tubulin antagonist.
357. The device of item 256 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG)1 an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
358. The device of item 256 wherein the agent is a VEGF inhibitor.
359. The device of item 256 wherein the agent is a vitamin D receptor agonist.
360. The device of item 256 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
361. The device of item 256 wherein the agent is AP-23573 (an mTOR inhibitor).
362. The device of item 256 wherein the agent is synthadotin (a tubulin antagonist).
363. The device of item 256 wherein the agent is S-0885 (a collagenase inhibitor).
364. The device of item 256 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
365. The device of item 256 wherein the agent is ixabepilone
(an epithilone).
366. The device of item 256 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
367. The device of item 256 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 368. The device of item 256 wherein the agent is ABT-518 (an angiogenesis inhibitor).
369. The device of item 256 wherein the agent is combretastatin (an angiogenesis inhibitor).
370. The device of item 256 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
371. The device of item 256 wherein the agent is SB-715992 (a kinesin antagonist).
372. The device of item 256 wherein the agent is temsirolimus (an mTOR inhibitor).
373. The device of item 256 wherein the agent is adalimumab (a TNFα antagonist).
374. The device of item 256, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
375. The device of item 256, further comprising a coating, wherein the coating comprises the anti-scarring agent.
376. The device of item 256, further comprising a coating, wherein the coating is disposed on a surface of the device.
377. The device of item 256, further comprising a coating, wherein the coating directly contacts the device.
378. The device of item 256, further comprising a coating, wherein the coating indirectly contacts the device.
379. The device of item 256, further comprising a coating, wherein the coating partially covers the device. 380. The device of item 256, further comprising a coating, wherein the coating completely covers the device.
381. The device of item 256, further comprising a coating, wherein the coating is a uniform coating.
382. The device of item 256, further comprising a coating, wherein the coating is a non-uniform coating.
383. The device of item 256, further comprising a coating, wherein the coating is a discontinuous coating.
384. The device of item 256, further comprising a coating, wherein the coating is a patterned coating.
385. The device of item 256, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
386. The device of item 256, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
387. The device of item 256, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
388. The device of item 256, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
389. The device of item 256, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
390. The device of item 256, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight. 391. The device of item 256, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
392. The device of item 256, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
393. The device of item 256, further comprising a coating, wherein the coating further comprises a polymer.
394. The device of item 256, further comprising a first coating having a first composition and the second coating having a second composition.
395. The device of item 256, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
396. The device of item 256, further comprising a polymer.
397. The device of item 256, further comprising a polymeric carrier.
398. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
399. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
400. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
401. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer. 402. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
403. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
404. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
405. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
406. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
407. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
408. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
409. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
410. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
411. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
412. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer. 413. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
414. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
415. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
416. The device of item 256, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
417. The device of item 256, further comprising a lubricious coating.
418. The device of item 256 wherein the anti-scarring agent is located within pores or holes of the device.
419. The device of item 256 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
420. The device of item 256, further comprising a second pharmaceutically active agent.
421. The device of item 256, further comprising an antiinflammatory agent.
422. The device of item 256, further comprising an agent that inhibits infection.
423. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
424. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is doxorubicin. 425. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
426. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
427. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
428. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
429. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
430. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
431. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is etoposide.
432. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
433. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
434. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
435. The device of item 256, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
436. The device of item 256, further comprising an antithrombotic agent. 437. The device of item 256, further comprising a visualization agent.
438. The device of item 256, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
439. The device of item 256, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
440. The device of item 256, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
441. The device of item 256, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
442. The device of item 256, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
443. The device of item 256, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
444. The device of item 256, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
445. The device of item 256, further comprising an echogenic material.
446. The device of item 256, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
447. The device of item 256 wherein the device is sterile. 448. The device of item 256 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
449. The device of item 256 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
450. The device of item 256 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
451. The device of item 256 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
452. The device of item 256 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
453. The device of item 256 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
454. The device of item 256 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
455. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
456. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 457. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
458. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
459. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
460. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
461. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
462. The device of item 256 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
463. The device of item 256 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
464. The device of item 256 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
465. The device of item 256 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
466. The device of item 256 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. 467. The device of item 256 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
468. The device of item 256 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
469. The device of item 256 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
470. The device of item 256 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
471. The device of item 256 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
472. The device of item 256 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
473. The device of item 256 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
474. The device of item 256 wherein the agent or the composition is affixed to the sensor.
475. The device of item 256 wherein the agent or the composition is covalently attached to the sensor. 476. The device of item 256 wherein the agent or the composition is non-covaiently attached to the sensor.
477. The device of item 256 further comprising a coating that absorbs the agent or the composition.
478. The device of item 256 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
479. The device of item 256 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
480. The device of item 256 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
481. The device of item 256 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
482. The device of item 256 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
483. The device of item 256 further comprising a pump that is linked to the sensor.
484. The device of any one of items 256-483 wherein the device is deliverable to the vascular system transluminal^ using a catheter on a stent platform.
485. The device of any one of items 256-483 wherein the device is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
486. The device of any one of items 256-483 wherein the device is an electrode composed of an analyte responsive enzyme. 487. The device of any one of items 256-483 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
488. The device of any one of items 256-483 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin.
489. A device, comprising a pressure or stress sensor and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
490. The device of item 489 wherein the agent is an adensosine A2A receptor antagonist.
491. The device of item 489 wherein the agent is an AKT inhibitor.
492. The device of item 489 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
493. The device of item 489 wherein the agent is an alpha 4 integrin antagonist.
494. The device of item 489 wherein the agent is an alpha 7 nicotinic receptor agonist.
495. The device of item 489 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
496. The device of item 489 wherein the agent is an apoptosis antagonist.
497. The device of item 489 wherein the agent is an apoptosis activator. 498. The device of item 489 wherein the agent is a beta 1 integrin antagonist.
499. The device of item 489 wherein the agent is a beta tubulin inhibitor.
500. The device of item 489 wherein the agent is a blocker of enzyme production in Hepatitis C.
501. The device of item 489 wherein the agent is a Bruton's tyrosine kinase inhibitor.
502. The device of item 489 wherein the agent is a calcineurin inhibitor.
503. The device of item 489 wherein the agent is a caspase 3 inhibitor.
504. The device of item 489 wherein the agent is a CC chemokine receptor antagonist.
505. The device of item 489 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
506. The device of item 489 wherein the agent is a cathepsin B inhibitor.
507. The device of item 489 wherein the agent is a cathepsin
K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
508. The device of item 489 wherein the agent is a cathepsin L inhibitor. 509. The device of item 489 wherein the agent is a CD40 antagonist.
510. The device of item 489 wherein the agent is a chemokine receptor agonist.
511. The device of item 489 wherein the agent is a chymase inhibitor.
512. The device of item 489 wherein the agent is a collagenase antagonist.
513. The device of item 489 wherein the agent is a CXCR antagonist.
514. The device of item 489 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
515. The device of item 489 wherein the agent is a cyclooxygenase 1 inhibitor.
516. The device of item 489 wherein the agent is a DHFR inhibitor.
517. The device of item 489 wherein the agent is a dual integrin inhibitor. 518. The device of item 489 wherein the agent is an elastase inhibitor.
519. The device of item 489 wherein the agent is an elongation factor-1 alpha inhibitor.
520. The device of item 489 wherein the agent is an endothelial growth factor antagonist.
521. The device of item 489 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
522. The device of item 489 wherein the agent is an endotoxin antagonist.
523. The device of item 489 wherein the agent is an epothilone and tubulin binder.
524. The device of item 489 wherein the agent is an estrogen receptor antagonist.
525. The device of item 489 wherein the agent is an FGF inhibitor.
526. The device of item 489 wherein the agent is a farnexyl transferase inhibitor. 527. The device of item 489 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
528. The device of item 489 wherein the agent is an FLT-3 kinase inhibitor.
529. The device of item 489 wherein the agent is an FGF receptor kinase inhibitor.
530. The device of item 489 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
531. The device of item 489 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
532. The device of item 489 wherein the agent is a histone deacetylase inhibitor.
533. The device of item 489 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
534. The device of item 489 wherein the agent is an ICAM inhibitor.
535. The device of item 489 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
536. The device of item 489 wherein the agent is an IL-2 inhibitor.
537. The device of item 489 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 538. The device of item 489 wherein the agent is an IMPDH (inosine monophosphate).
539. The device of item 489 wherein the agent is an integrin antagonist.
540. The device of item 489 wherein the agent is an interleukin antagonist.
541. The device of item 489 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
542. The device of item 489 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
543. The device of item 489 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
544. The device of item 489 wherein the agent a JAK3 enzyme inhibitor.
545. The device of item 489 wherein the agent is a JNK inhibitor.
546. The device of item 489 wherein the agent is a kinase inhibitor.
547. The device of item 489 wherein the agent is kinesin antagonist.
548. The device of item 489 wherein the agent is a kinesin antagonist.
549. The device of item 489 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
550. The device of item 489 wherein the agent is an MAP kinase inhibitor.
551. The device of item 489 wherein the agent is a matrix metalloproteinase inhibitor.
552. The device of item 489 wherein the agent is an MCP- CCR2 inhibitor.
553. The device of item 489 wherein the agent is an mTOR inhibitor.
554. The device of item 489 wherein the agent is an mTOR kinase inhibitor.
555. The device of item 489 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
556. The device of item 489 wherein the agent is an MIF inhibitor.
557. The device of item 489 wherein the agent is an MMP inhibitor.
558. The device of item 489 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL- 105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
559. The device of item 489 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-
576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
560. The device of item 489 wherein the agent is a nitric oxide agonist.
561. The device of item 489 wherein the agent is an ornithine decarboxylase inhibitor.
562. The device of item 489 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
563. The device of item 489 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
564. The device of item 489 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
565. The device of item 489 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
566. The device of item 489 wherein the agent is a phosphatase inhibitor.
567. The device of item 489 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (In2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
568. The device of item 489 wherein the agent is a PKC inhibitor.
569. The device of item 489 wherein the agent is a platelet activating factor antagonist.
570. The device of item 489 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
571. The device of item 489 wherein the agent is a prolyl hydroxylase inhibitor.
572. The device of item 489 wherein the agent is a polymorphonuclear neutrophil inhibitor.
573. The device of item 489 wherein the agent is a protein kinase B inhibitor.
574. The device of item 489 wherein the agent is a protein kinase C stimulant.
575. The device of item 489 wherein the agent is a purine nucleoside analogue.
576. The device of item 489 wherein the agent is a purinoreceptor P2X antagonist. 577. The device of item 489 wherein the agent is a Raf kinase inhibitor.
578. The device of item 489 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
579. The device of item 489 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
580. The device of item 489 wherein the agent is an SDF-1 antagonist.
581. The device of item 489 wherein the agent is a sheddase inhibitor.
582. The device of item 489 wherein the agent is an SRC inhibitor.
583. The device of item 489 wherein the agent is a stromelysin inhibitor.
584. The device of item 489 wherein the agent is an Syk kinase inhibitor.
585. The device of item 489 wherein the agent is a telomerase inhibitor.
586. The device of item 489 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-
8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG)1 TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 587. The device of item 489 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
588. The device of item 489 wherein the agent is a Toll receptor inhibitor.
589. The device of item 489 wherein the agent is a tubulin antagonist.
590. The device of item 489 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
591. The device of item 489 wherein the agent is a VEGF inhibitor.
592. The device of item 489 wherein the agent is a vitamin D receptor agonist.
593. The device of item 489 wherein the agent is ZD-6474 (an angiogenesis inhibitor). 594. The device of item 489 wherein the agent is AP-23573 (an mTOR inhibitor).
595. The device of item 489 wherein the agent is synthadotin (a tubulin antagonist).
596. The device of item 489 wherein the agent is S-0885 (a collagenase inhibitor).
597. The device of item 489 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
598. The device of item 489 wherein the agent is ixabepilone (an epithilone).
599. The device of item 489 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
600. The device of item 489 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
601. The device of item 489 wherein the agent is ABT-518 (an angiogenesis inhibitor).
602. The device of item 489 wherein the agent is combretastatin (an angiogenesis inhibitor).
603. The device of item 489 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
604. The device of item 489 wherein the agent is SB-715992 (a kinesin antagonist).
605. The device of item 489 wherein the agent is temsirolimus (an mTOR inhibitor). 606. The device of item 489 wherein the agent is adalimumab (a TNFα antagonist).
607. The device of item 489, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
608. The device of item 489, further comprising a coating, wherein the coating comprises the anti-scarring agent.
609. The device of item 489, further comprising a coating, wherein the coating is disposed on a surface of the device.
610. The device of item 489, further comprising a coating, wherein the coating directly contacts the device.
611. The device of item 489, further comprising a coating, wherein the coating indirectly contacts the device.
612. The device of item 489, further comprising a coating, wherein the coating partially covers the device.
613. The device of item 489, further comprising a coating, wherein the coating completely covers the device.
614. The device of item 489, further comprising a coating, wherein the coating is a uniform coating.
615. The device of item 489, further comprising a coating, wherein the coating is a non-uniform coating.
616. The device of item 489, further comprising a coating, wherein the coating is a discontinuous coating.
617. The device of item 489, further comprising a coating, wherein the coating is a patterned coating. 618. The device of item 489, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
619. The device of item 489, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
620. The device of item 489, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
621. The device of item 489, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
622. The device of item 489, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
623. The device of item 489, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
624. The device of item 489, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
625. The device of item 489, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
626. The device of item 489, further comprising a coating, wherein the coating further comprises a polymer.
627. The device of item 489, further comprising a first coating having a first composition and the second coating having a second composition. 628. The device of item 489, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
629. The device of item 489, further comprising a polymer.
630. The device of item 489, further comprising a polymeric carrier.
631. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
632. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
633. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
634. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
635. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
636. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
637. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
638. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains. 639. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
640. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
641. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
642. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
643. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
644. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
645. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
646. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
647. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
648. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
649. The device of item 489, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer. 650. The device of item 489, further comprising a lubricious coating.
651. The device of item 489 wherein the anti-scarring agent is located within pores or holes of the device.
652. The device of item 489 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
653. The device of item 489, further comprising a second pharmaceutically active agent.
654. The device of item 489, further comprising an anti- inflammatory agent.
655. The device of item 489, further comprising an agent that inhibits infection.
656. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
657. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
658. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
659. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
660. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
661. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. 662. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
663. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
664. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is etoposide.
665. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
666. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
667. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
668. The device of item 489, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
669. The device of item 489, further comprising an antithrombotic agent.
670. The device of item 489, further comprising a visualization agent.
671. The device of item 489, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
672. The device of item 489, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium. 673. The device of item 489, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
674. The device of item 489, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
675. The device of item 489, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
676. The device of item 489, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
677. The device of item 489, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
678. The device of item 489, further comprising an echogenic material.
679. The device of item 489, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
680. The device of item 489 wherein the device is sterile.
681. The device of item 489 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
682. The device of item 489 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
683. The device of item 489 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. 684. The device of item 489 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
685. The device of item 489 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
686. The device of item 489 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
687. The device of item 489 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
688. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
689. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
690. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
691. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
692. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 693. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
694. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
695. The device of item 489 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
696. The device of item 489 wherein the device comprises about 0.01 μm to about 10 μg of the anti-scarring agent.
697. The device of item 489 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
698. The device of item 489 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
699. The device of item 489 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
700. The device of item 489 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
701. The device of item 489 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
702. The device of item 489 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 703. The device of item 489 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
704. The device of item 489 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
705. The device of item 489 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
706. The device of item 489 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
707. The device of item 489 wherein the agent or the composition is affixed to the sensor.
708. The device of item 489 wherein the agent or the composition is covalently attached to the sensor.
709. The device of item 489 wherein the agent or the composition is non-covalently attached to the sensor.
710. The device of item 489 further comprising a coating that absorbs the agent or the composition.
711. The device of item 489 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
712. The device of item 489 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition. 713. The device of item 489 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
714. The device of item 489 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
715. The device of item 489 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
716. The device of item 489 further comprising a pump that is linked to the sensor.
717. The device of any one of items 489-716 wherein the device monitors blood pressure.
718. The device of any one of items 489-716 wherein the device monitors fluid flow.
719. The device of any one of items 489-716 wherein the device monitors pressure within an aneurysm sac.
720. The device of any one of items 489-716 wherein the device monitors intracranial pressure.
721. The device of any one of items 489-716 wherein the device monitors mechanical pressure associated with a bone fracture.
722. The device of any one of items 489-716 wherein the device monitors barometric pressure.
723. The device of any one of items 489-716 wherein the device monitors eye tremors.
724. The device of any one of items 489-716 wherein the device monitors the depth of a corneal implant. 725. The device of any one of items 489-716 wherein the device monitors intraocular pressure.
726. The device of any one of items 489-716 wherein the device is a passive sensor with an inductor-capacitor circuit.
727. The device of any one of items 489-716 wherein the device is a self-powered strain sensing system.
728. The device of any one of items 489-716 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage.
729. A device, comprising a cardiac sensor and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
730. The device of item 729 wherein the agent is an adensosine A2A receptor antagonist.
731. The device of item 729 wherein the agent is an AKT inhibitor.
732. The device of item 729 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
733. The device of item 729 wherein the agent is an alpha 4 integrin antagonist.
734. The device of item 729 wherein the agent is an alpha 7 nicotinic receptor agonist.
735. The device of item 729 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant
Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005
(GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732
(Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA)1 NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-
23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
736. The device of item 729 wherein the agent is an apoptosis antagonist. 737. The device of item 729 wherein the agent is an apoptosis activator.
738. The device of item 729 wherein the agent is a beta 1 integrin antagonist.
739. The device of item 729 wherein the agent is a beta tubulin inhibitor.
740. The device of item 729 wherein the agent is a blocker of enzyme production in Hepatitis C.
741. The device of item 729 wherein the agent is a Bruton's tyrosine kinase inhibitor.
742. The device of item 729 wherein the agent is a calcineurin inhibitor.
743. The device of item 729 wherein the agent is a caspase 3 inhibitor.
744. The device of item 729 wherein the agent is a CC chemokine receptor antagonist.
745. The device of item 729 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
746. The device of item 729 wherein the agent is a cathepsin
B inhibitor.
747. The device of item 729 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 748. The device of item 729 wherein the agent is a cathepsin L inhibitor.
749. The device of item 729 wherein the agent is a CD40 antagonist.
750. The device of item 729 wherein the agent is a chemokine receptor agonist.
751. The device of item 729 wherein the agent is a chymase inhibitor.
752. The device of item 729 wherein the agent is a collagenase antagonist.
753. The device of item 729 wherein the agent is a CXCR antagonist.
754. The device of item 729 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
755. The device of item 729 wherein the agent is a cyclooxygenase 1 inhibitor.
756. The device of item 729 wherein the agent is a DHFR inhibitor. 757. The device of item 729 wherein the agent is a dual integrin inhibitor.
758. The device of item 729 wherein the agent is an elastase inhibitor.
759. The device of item 729 wherein the agent is an elongation factor-1 alpha inhibitor.
760. The device of item 729 wherein the agent is an endothelial growth factor antagonist.
761. The device of item 729 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI
Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
762. The device of item 729 wherein the agent is an endotoxin antagonist.
763. The device of item 729 wherein the agent is an epothilone and tubulin binder.
764. The device of item 729 wherein the agent is an estrogen receptor antagonist.
765. The device of item 729 wherein the agent is an FGF inhibitor. 766. The device of item 729 wherein the agent is a farnexyl transferase inhibitor.
767. The device of item 729 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
768. The device of item 729 wherein the agent is an FLT-3 kinase inhibitor.
769. The device of item 729 wherein the agent is an FGF receptor kinase inhibitor.
770. The device of item 729 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11 -9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
771. The device of item 729 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005
(Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
772. The device of item 729 wherein the agent is a histone deacetylase inhibitor. 773. The device of item 729 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
774. The device of item 729 wherein the agent is an ICAM inhibitor.
775. The device of item 729 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
776. The device of item 729 wherein the agent is an IL-2 inhibitor.
777. The device of item 729 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
778. The device of item 729 wherein the agent is an IMPDH (inosine monophosphate).
779. The device of item 729 wherein the agent is an integrin antagonist.
780. The device of item 729 wherein the agent is an interleukin antagonist.
781. The device of item 729 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
782. The device of item 729 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
783. The device of item 729 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
784. The device of item 729 wherein the agent a JAK3 enzyme inhibitor.
785. The device of item 729 wherein the agent is a JNK inhibitor.
786. The device of item 729 wherein the agent is a kinase inhibitor.
787. The device of item 729 wherein the agent is kinesin antagonist.
788. The device of item 729 wherein the agent is a kinesin antagonist. 789. The device of item 729 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
790. The device of item 729 wherein the agent is an MAP kinase inhibitor.
791. The device of item 729 wherein the agent is a matrix metalloproteinase inhibitor.
792. The device of item 729 wherein the agent is an MCP- CCR2 inhibitor.
793. The device of item 729 wherein the agent is an mTOR inhibitor.
794. The device of item 729 wherein the agent is an mTOR kinase inhibitor.
795. The device of item 729 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
796. The device of item 729 wherein the agent is an MIF inhibitor.
797. The device of item 729 wherein the agent is an MMP inhibitor.
798. The device of item 729 wherein the agent is a neurokinin
(NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL- 105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
799. The device of item 729 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
800. The device of item 729 wherein the agent is a nitric oxide agonist.
801. The device of item 729 wherein the agent is an ornithine decarboxylase inhibitor.
802. The device of item 729 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca),
JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
803. The device of item 729 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
804. The device of item 729 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
805. The device of item 729 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
806. The device of item 729 wherein the agent is a phosphatase inhibitor.
807. The device of item 729 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase 111 inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
808. The device of item 729 wherein the agent is a PKC inhibitor.
809. The device of item 729 wherein the agent is a platelet activating factor antagonist.
810. The device of item 729 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
811. The device of item 729 wherein the agent is a prolyl hydroxylase inhibitor.
812. The device of item 729 wherein the agent is a polymorphonuclear neutrophil inhibitor.
813. The device of item 729 wherein the agent is a protein kinase B inhibitor.
814. The device of item 729 wherein the agent is a protein kinase C stimulant.
815. The device of item 729 wherein the agent is a purine nucleoside analogue. 816. The device of item 729 wherein the agent is a purinoreceptor P2X antagonist.
817. The device of item 729 wherein the agent is a Raf kinase inhibitor.
818. The device of item 729 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
819. The device of item 729 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
820. The device of item 729 wherein the agent is an SDF-1 antagonist.
821. The device of item 729 wherein the agent is a sheddase inhibitor.
822. The device of item 729 wherein the agent is an SRC inhibitor.
823. The device of item 729 wherein the agent is a stromelysin inhibitor.
824. The device of item 729 wherein the agent is an Syk kinase inhibitor.
825. The device of item 729 wherein the agent is a telomerase inhibitor.
826. The device of item 729 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
827. The device of item 729 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
828. The device of item 729 wherein the agent is a Toll receptor inhibitor.
829. The device of item 729 wherein the agent is a tubulin antagonist.
830. The device of item 729 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
831. The device of item 729 wherein the agent is a VEGF inhibitor.
832. The device of item 729 wherein the agent is a vitamin D receptor agonist. 833. The device of item 729 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
834. The device of item 729 wherein the agent is AP-23573 (an mTOR inhibitor).
835. The device of item 729 wherein the agent is synthadotin
(a tubulin antagonist).
836. The device of item 729 wherein the agent is S-0885 (a collagenase inhibitor).
837. The device of item 729 wherein the agent is aplidine (an elongation factor- 1 alpha inhibitor).
838. The device of item 729 wherein the agent is ixabepilone (an epithilone).
839. The device of item 729 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
840. The device of item 729 wherein the agent is SB-2723005
(an angiogenesis inhibitor).
841. The device of item 729 wherein the agent is ABT-518 (an angiogenesis inhibitor).
842. The device of item 729 wherein the agent is combretastatin (an angiogenesis inhibitor).
843. The device of item 729 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
844. The device of item 729 wherein the agent is SB-715992 (a kinesin antagonist). 845. The device of item 729 wherein the agent is temsirolimus (an mTOR inhibitor).
846. The device of item 729 wherein the agent is adalimumab (a TN Fa antagonist).
847. The device of item 729, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
848. The device of item 729, further comprising a coating, wherein the coating comprises the anti-scarring agent.
849. The device of item 729, further comprising a coating, wherein the coating is disposed on a surface of the device.
850. The device of item 729, further comprising a coating, wherein the coating directly contacts the device.
851. The device of item 729, further comprising a coating, wherein the coating indirectly contacts the device.
852. The device of item 729, further comprising a coating, wherein the coating partially covers the device.
853. The device of item 729, further comprising a coating, wherein the coating completely covers the device.
854. The device of item 729, further comprising a coating, wherein the coating is a uniform coating.
855. The device of item 729, further comprising a coating, wherein the coating is a non-uniform coating.
856. The device of item 729, further comprising a coating, wherein the coating is a discontinuous coating. 857. The device of item 729, further comprising a coating, wherein the coating is a patterned coating.
858. The device of item 729, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
859. The device of item 729, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
860. The device of item 729, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
861. The device of item 729, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
862. The device of item 729, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
863. The device of item 729, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
864. The device of item 729, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
865. The device of item 729, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
866. The device of item 729, further comprising a coating, wherein the coating further comprises a polymer. 867. The device of item 729, further comprising a first coating having a first composition and the second coating having a second composition.
868. The device of item 729, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
869. The device of item 729, further comprising a polymer.
870. The device of item 729, further comprising a polymeric carrier.
871. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
872. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
873. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
874. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
875. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
876. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
877. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer. 878. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
879. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
880. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
881. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
882. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
883. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
884. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
885. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
886. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
887. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
888. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 889. The device of item 729, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
890. The device of item 729, further comprising a lubricious coating.
891. The device of item 729 wherein the anti-scarring agent is located within pores or holes of the device.
892. The device of item 729 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
893. The device of item 729, further comprising a second pharmaceutically active agent.
894. The device of item 729, further comprising an antiinflammatory agent.
895. The device of item 729, further comprising an agent that inhibits infection.
896. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
897. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
898. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
899. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
900. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU). 901. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
902. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
903. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
904. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is etoposide.
905. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
906. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
907. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
908. The device of item 729, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
909. The device of item 729, further comprising an antithrombotic agent.
910. The device of item 729, further comprising a visualization agent.
911. The device of item 729, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 912. The device of item 729, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
913. The device of item 729, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
914. The device of item 729, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
915. The device of item 729, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
916. The device of item 729, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
917. The device of item 729, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
918. The device of item 729, further comprising an echogenic material.
919. The device of item 729, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
920. The device of item 729 wherein the device is sterile.
921. The device of item 729 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
922. The device of item 729 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device. 923. The device of item 729 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
924. The device of item 729 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
925. The device of item 729 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
926. The device of item 729 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
927. The device of item 729 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
928. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
929. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
930. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
931. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate. 932. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
933. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
934. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
935. The device of item 729 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
936. The device of item 729 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
937. The device of item 729 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
938. The device of item 729 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
939. The device of item 729 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
940. The device of item 729 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
941. The device of item 729 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 942. The device of item 729 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
943. The device of item 729 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
944. The device of item 729 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
945. The device of item 729 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
946. The device of item 729 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
947. The device of item 729 wherein the agent or the composition is affixed to the sensor.
948. The device of item 729 wherein the agent or the composition is covalently attached to the sensor.
949. The device of item 729 wherein the agent or the composition is non-covalently attached to the sensor.
950. The device of item 729 further comprising a coating that absorbs the agent or the composition.
951. The device of item 729 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition. 952. The device of item 729 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
953. The device of item 729 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
954. The device of item 729 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
955. The device of item 729 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
956. The device of item 729 further comprising a pump that is linked to the sensor.
957. The device of any one of items 729-956 wherein the device monitors cardiac output.
958. The device of any one of items 729-956 wherein the device monitors ejection fraction.
959. The device of any one of items 729-956 wherein the device monitors blood pressure in a heart chamber.
960. The device of any one of items 729-956 wherein the device monitors ventricular wall motions.
961. The device of any one of items 729-956 wherein the device monitors blood flow to a transplanted organ.
962. The device of any one of items 729-956 wherein the device monitors heart rate.
963. A device, comprising a respiratory sensor and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
964. The device of item 963 wherein the agent is an adensosine A2A receptor antagonist.
965. The device of item 963 wherein the agent is an AKT inhibitor.
966. The device of item 963 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
967. The device of item 963 wherein the agent is an alpha 4 integrin antagonist.
968. The device of item 963 wherein the agent is an alpha 7 nicotinic receptor agonist.
969. The device of item 963 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GiaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
970. The device of item 963 wherein the agent is an apoptosis antagonist.
971. The device of item 963 wherein the agent is an apoptosis activator.
972. The device of item 963 wherein the agent is a beta 1 integrin antagonist.
973. The device of item 963 wherein the agent is a beta tubulin inhibitor.
974. The device of item 963 wherein the agent is a blocker of enzyme production in Hepatitis C.
975. The device of item 963 wherein the agent is a Bruton's tyrosine kinase inhibitor. 976. The device of item 963 wherein the agent is a calcineurin inhibitor.
977. The device of item 963 wherein the agent is a caspase 3 inhibitor.
978. The device of item 963 wherein the agent is a CC chemokine receptor antagonist.
979. The device of item 963 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
980. The device of item 963 wherein the agent is a cathepsin
B inhibitor.
981. The device of item 963 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
982. The device of item 963 wherein the agent is a cathepsin L inhibitor.
983. The device of item 963 wherein the agent is a CD40 antagonist.
984. The device of item 963 wherein the agent is a chemokine receptor agonist.
985. The device of item 963 wherein the agent is a chymase inhibitor.
986. The device of item 963 wherein the agent is a collagenase antagonist. 987. The device of item 963 wherein the agent is a CXCR antagonist.
988. The device of item 963 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
989. The device of item 963 wherein the agent is a cyclooxygenase 1 inhibitor.
990. The device of item 963 wherein the agent is a DHFR inhibitor.
991. The device of item 963 wherein the agent is a dual integrin inhibitor.
992. The device of item 963 wherein the agent is an elastase inhibitor.
993. The device of item 963 wherein the agent is an elongation factor-1 alpha inhibitor.
994. The device of item 963 wherein the agent is an endothelial growth factor antagonist.
995. The device of item 963 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI
Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
996. The device of item 963 wherein the agent is an endotoxin antagonist.
997. The device of item 963 wherein the agent is an epothilone and tubulin binder.
998. The device of item 963 wherein the agent is an estrogen receptor antagonist.
999. The device of item 963 wherein the agent is an FGF inhibitor.
1000. The device of item 963 wherein the agent is a farnexyl transferase inhibitor.
1001. The device of item 963 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1002. The device of item 963 wherein the agent is an FLT-3 kinase inhibitor. 1003. The device of item 963 wherein the agent is an FGF receptor kinase inhibitor.
1004. The device of item 963 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1005. The device of item 963 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005
(Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
1006. The device of item 963 wherein the agent is a histone deacetylase inhibitor.
1007. The device of item 963 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
1008. The device of item 963 wherein the agent is an ICAM inhibitor.
1009. The device of item 963 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174
(Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 1010. The device of item 963 wherein the agent is an IL-2 inhibitor.
1011. The device of item 963 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1012. The device of item 963 wherein the agent is an IMPDH (inosine monophosphate).
1013. The device of item 963 wherein the agent is an integrin antagonist.
1014. The device of item 963 wherein the agent is an interleukin antagonist.
1015. The device of item 963 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 1016. The device of item 963 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1017. The device of item 963 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1018. The device of item 963 wherein the agent a JAK3 enzyme inhibitor.
1019. The device of item 963 wherein the agent is a JNK inhibitor.
1020. The device of item 963 wherein the agent is a kinase inhibitor.
1021. The device of item 963 wherein the agent is kinesin antagonist.
1022. The device of item 963 wherein the agent is a kinesin antagonist.
1023. The device of item 963 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1024. The device of item 963 wherein the agent is an MAP kinase inhibitor.
1025. The device of item 963 wherein the agent is a matrix metalloproteinase inhibitor.
1026. The device of item 963 wherein the agent is an MCP- CCR2 inhibitor.
1027. The device of item 963 wherein the agent is an mTOR inhibitor.
1028. The device of item 963 wherein the agent is an mTOR kinase inhibitor.
1029. The device of item 963 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1030. The device of item 963 wherein the agent is an MIF inhibitor. 1031. The device of item 963 wherein the agent is an MMP inhibitor.
1032. The device of item 963 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-
105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1033. The device of item 963 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1034. The device of item 963 wherein the agent is a nitric oxide agonist.
1035. The device of item 963 wherein the agent is an ornithine decarboxylase inhibitor.
1036. The device of item 963 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1037. The device of item 963 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
1038. The device of item 963 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1039. The device of item 963 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1040. The device of item 963 wherein the agent is a phosphatase inhibitor.
1041. The device of item 963 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1042. The device of item 963 wherein the agent is a PKC inhibitor. 1043. The device of item 963 wherein the agent is a platelet activating factor antagonist.
1044. The device of item 963 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1045. The device of item 963 wherein the agent is a prolyl hydroxylase inhibitor.
1046. The device of item 963 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1047. The device of item 963 wherein the agent is a protein kinase B inhibitor.
1048. The device of item 963 wherein the agent is a protein kinase C stimulant.
1049. The device of item 963 wherein the agent is a purine nucleoside analogue.
1050. The device of item 963 wherein the agent is a purinoreceptor P2X antagonist.
1051. The device of item 963 wherein the agent is a Raf kinase inhibitor.
1052. The device of item 963 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
1053. The device of item 963 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1054. The device of item 963 wherein the agent is an SDF-1 antagonist. 1055. The device of item 963 wherein the agent is a sheddase inhibitor.
1056. The device of item 963 wherein the agent is an SRC inhibitor.
1057. The device of item 963 wherein the agent is a stromelysin inhibitor.
1058. The device of item 963 wherein the agent is an Syk kinase inhibitor.
1059. The device of item 963 wherein the agent is a telomerase inhibitor.
1060. The device of item 963 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1061. The device of item 963 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1062. The device of item 963 wherein the agent is a Toll receptor inhibitor. 1063. The device of item 963 wherein the agent is a tubulin antagonist.
1064. The device of item 963 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian
Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1065. The device of item 963 wherein the agent is a VEGF inhibitor.
1066. The device of item 963 wherein the agent is a vitamin D receptor agonist.
1067. The device of item 963 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
1068. The device of item 963 wherein the agent is AP-23573 (an mTOR inhibitor).
1069. The device of item 963 wherein the agent is synthadotin (a tubulin antagonist).
1070. The device of item 963 wherein the agent is S-0885 (a collagenase inhibitor).
1071. The device of item 963 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor). 1072. The device of item 963 wherein the agent is ixabepilone (an epithilone).
1073. The device of item 963 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1074. The device of item 963 wherein the agent is SB-2723005
(an angiogenesis inhibitor).
1075. The device of item 963 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1076. The device of item 963 wherein the agent is combretastatin (an angiogenesis inhibitor).
1077. The device of item 963 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1078. The device of item 963 wherein the agent is SB-715992 (a kinesin antagonist).
1079. The device of item 963 wherein the agent is temsirolimus
(an mTOR inhibitor).
1080. The device of item 963 wherein the agent is adalimumab (a TN Fa antagonist).
1081. The device of item 963, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
1082. The device of item 963, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1083. The device of item 963, further comprising a coating, wherein the coating is disposed on a surface of the device. 1084. The device of item 963, further comprising a coating, wherein the coating directly contacts the device.
1085. The device of item 963, further comprising a coating, wherein the coating indirectly contacts the device.
1086. The device of item 963, further comprising a coating, wherein the coating partially covers the device.
1087. The device of item 963, further comprising a coating, wherein the coating completely covers the device.
1088. The device of item 963, further comprising a coating, wherein the coating is a uniform coating.
1089. The device of item 963, further comprising a coating, wherein the coating is a non-uniform coating.
1090. The device of item 963, further comprising a coating, wherein the coating is a discontinuous coating.
1091. The device of item 963, further comprising a coating, wherein the coating is a patterned coating.
1092. The device of item 963, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
1093. The device of item 963, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
1094. The device of item 963, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1095. The device of item 963, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year. 1096. The device of item 963, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1097. The device of item 963, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
1098. The device of item 963, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1099. The device of item 963, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1100. The device of item 963, further comprising a coating, wherein the coating further comprises a polymer.
1101. The device of item 963, further comprising a first coating having a first composition and the second coating having a second composition.
1102. The device of item 963, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1103. The device of item 963, further comprising a polymer.
1104. The device of item 963, further comprising a polymeric carrier.
1105. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer. 1106. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1107. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1108. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1109. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1110. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1111. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1112. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1113. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1114. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1115. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1116. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel. 1117. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1118. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1119. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1120. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1121. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1122. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1123. The device of item 963, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1124. The device of item 963, further comprising a lubricious coating.
1125. The device of item 963 wherein the anti-scarring agent is located within pores or holes of the device.
1126. The device of item 963 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1127. The device of item 963, further comprising a second pharmaceutically active agent.
1128. The device of item 963, further comprising an anti- inflammatory agent. 1129. The device of item 963, further comprising an agent that inhibits infection.
1130. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1131. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1132. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1133. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1134. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1135. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1136. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1137. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1138. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1139. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1140. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 1141. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1142. The device of item 963, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1143. The device of item 963, further comprising an antithrombotic agent.
1144. The device of item 963, further comprising a visualization agent.
1145. The device of item 963, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1146. The device of item 963, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1147. The device of item 963, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1148. The device of item 963, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1149. The device of item 963, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1150. The device of item 963, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1151. The device of item 963, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant. 1152. The device of item 963, further comprising an echogenic material.
1153. The device of item 963, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1154. The device of item 963 wherein the device is sterile.
1155. The device of item 963 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1156. The device of item 963 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
1157. The device of item 963 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1158. The device of item 963 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1159. The device of item 963 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1160. The device of item 963 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1161. The device of item 963 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue. 1162. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1163. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1164. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1165. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1166. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1167. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1168. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1169. The device of item 963 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1170. The device of item 963 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent. 1171. The device of item 963 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
1172. The device of item 963 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1173. The device of item 963 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1174. The device of item 963 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1175. The device of item 963 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1176. The device of item 963 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1177. The device of item 963 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1178. The device of item 963 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1179. The device of item 963 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1180. The device of item 963 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1181. The device of item 963 wherein the agent or the composition is affixed to the sensor.
1182. The device of item 963 wherein the agent or the composition is covalently attached to the sensor.
1183. The device of item 963 wherein the agent or the composition is non-covalently attached to the sensor.
1184. The device of item 963 further comprising a coating that absorbs the agent or the composition.
1185. The device of item 963 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
1186. The device of item 963 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
1187. The device of item 963 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
1188. The device of item 963 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
1189. The device of item 963 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
1190. The device of item 963 further comprising a pump that is linked to the sensor.
1191. The device of any one of items 963-1190 wherein the device monitors pulmonary functions. 1192. A device, comprising an auditory sensor and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1193. The device of item 1192 wherein the agent is an adensosine A2A receptor antagonist.
1194. The device of item 1192 wherein the agent is an AKT inhibitor.
1195. The device of item 1192 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects
No. 5754 (Merck KgaA).
1196. The device of item 1192 wherein the agent is an alpha 4 integrin antagonist.
1197. The device of item 1192 wherein the agent is an alpha 7 nicotinic receptor agonist.
1198. The device of item 1192 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), • mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005
(GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1199. The device of item 1192 wherein the agent is an apoptosis antagonist.
1200. The device of item 1192 wherein the agent is an apoptosis activator.
1201. The device of item 1192 wherein the agent is a beta 1 integrin antagonist.
1202. The device of item 1192 wherein the agent is a beta tubulin inhibitor.
1203. The device of item 1192 wherein the agent is a blocker of enzyme production in Hepatitis C. 1204. The device of item 1192 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1205. The device of item 1192 wherein the agent is a calcineurin inhibitor.
1206. The device of item 1192 wherein the agent is a caspase 3 inhibitor.
1207. The device of item 1192 wherein the agent is a CC chemokine receptor antagonist.
1208. The device of item 1192 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin,
KRX-0403, and an analogue or derivative thereof.
1209. The device of item 1192 wherein the agent is a cathepsin B inhibitor.
1210. The device of item 1192 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline),
INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
1211. The device of item 1192 wherein the agent is a cathepsin L inhibitor.
1212. The device of item 1192 wherein the agent is a CD40 antagonist.
1213. The device of item 1192 wherein the agent is a chemokine receptor agonist.
1214. The device of item 1192 wherein the agent is a chymase inhibitor. 1215. The device of item 1192 wherein the agent is a collagenase antagonist.
1216. The device of item 1192 wherein the agent is a CXCR antagonist.
1217. The device of item 1192 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
1218. The device of item 1192 wherein the agent is a cyclooxygenase 1 inhibitor.
1219. The device of item 1192 wherein the agent is a DHFR inhibitor.
1220. The device of item 1192 wherein the agent is a dual integrin inhibitor.
1221. The device of item 1192 wherein the agent is an elastase inhibitor.
1222. The device of item 1192 wherein the agent is an elongation factor-1 alpha inhibitor.
1223. The device of item 1192 wherein the agent is an endothelial growth factor antagonist. 1224. The device of item 1192 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1225. The device of item 1192 wherein the agent is an endotoxin antagonist.
1226. The device of item 1192 wherein the agent is an epothilone and tubulin binder.
1227. The device of item 1192 wherein the agent is an estrogen receptor antagonist.
1228. The device of item 1192 wherein the agent is an FGF inhibitor.
1229. The device of item 1192 wherein the agent is a farnexyl transferase inhibitor.
1230. The device of item 1192 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme),
Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof. 1231. The device of item 1192 wherein the agent is an FLT-3 kinase inhibitor.
1232. The device of item 1192 wherein the agent is an FGF receptor kinase inhibitor.
1233. The device of item 1192 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1234. The device of item 1192 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
1235. The device of item 1192 wherein the agent is a histone deacetylase inhibitor.
1236. The device of item 1192 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
1237. The device of item 1192 wherein the agent is an ICAM inhibitor. 1238. The device of item 1192 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1239. The device of item 1192 wherein the agent is an IL-2 inhibitor.
1240. The device of item 1192 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1241. The device of item 1192 wherein the agent is an IMPDH (inosine monophosphate).
1242. The device of item 1192 wherein the agent is an integrin antagonist. 1243. The device of item 1192 wherein the agent is an interleukin antagonist.
1244. The device of item 1192 wherein the agent is an inhibitor of type IK receptor tyrosine kinase.
1245. The device of item 1192 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1246. The device of item 1192 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1247. The device of item 1192 wherein the agent a JAK3 enzyme inhibitor.
1248. The device of item 1192 wherein the agent is a JNK inhibitor.
1249. The device of item 1192 wherein the agent is a kinase inhibitor.
1250. The device of item 1192 wherein the agent is kinesin antagonist.
1251. The device of item 1192 wherein the agent is a kinesin antagonist.
1252. The device of item 1192 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1253. The device of item 1192 wherein the agent is an MAP kinase inhibitor.
1254. The device of item 1192 wherein the agent is a matrix metalloproteinase inhibitor.
1255. The device of item 1192 wherein the agent is an MCP- CCR2 inhibitor.
1256. The device of item 1192 wherein the agent is an mTOR inhibitor.
1257. The device of item 1192 wherein the agent is an mTOR kinase inhibitor.
1258. The device of item 1192 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1259. The device of item 1192 wherein the agent is an MIF inhibitor.
1260. The device of item 1192 wherein the agent is an MMP inhibitor.
1261. The device of item 1192 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1262. The device of item 1192 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1263. The device of item 1192 wherein the agent is a nitric oxide agonist. 1264. The device of item 1192 wherein the agent is an ornithine decarboxylase inhibitor.
1265. The device of item 1192 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1266. The device of item 1192 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
1267. The device of item 1192 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1268. The device of item 1192 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1269. The device of item 1192 wherein the agent is a phosphatase inhibitor.
1270. The device of item 1192 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1271. The device of item 1192 wherein the agent is a PKC inhibitor.
1272. The device of item 1192 wherein the agent is a platelet activating factor antagonist.
1273. The device of item 1192 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1274. The device of item 1192 wherein the agent is a prolyl hydroxylase inhibitor.
1275. The device of item 1192 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1276. The device of item 1192 wherein the agent is a protein kinase B inhibitor.
1277. The device of item 1192 wherein the agent is a protein kinase C stimulant.
1278. The device of item 1192 wherein the agent is a purine nucleoside analogue.
1279. The device of item 1192 wherein the agent is a purinoreceptor P2X antagonist.
1280. The device of item 1192 wherein the agent is a Raf kinase inhibitor.
1281. The device of item 1192 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 1282. The device of item 1192 wherein the agent is a ribonυcleoside triphosphate reductase inhibitor.
1283. The device of item 1192 wherein the agent is an SDF-1 antagonist.
1284. The device of item 1192 wherein the agent is a sheddase inhibitor.
1285. The device of item 1192 wherein the agent is an SRC inhibitor.
1286. The device of item 1192 wherein the agent is a stromelysin inhibitor.
1287. The device of item 1192 wherein the agent is an Syk kinase inhibitor.
1288. The device of item 1192 wherein the agent is a telomerase inhibitor.
1289. The device of item 1192 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG)1 TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1290. The device of item 1192 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1291. The device of item 1192 wherein the agent is a Toll receptor inhibitor.
1292. The device of item 1192 wherein the agent is a tubulin antagonist.
1293. The device of item 1192 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1294. The device of item 1192 wherein the agent is a VEGF inhibitor.
1295. The device of item 1192 wherein the agent is a vitamin D receptor agonist.
1296. The device of item 1192 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
1297. The device of item 1192 wherein the agent is AP-23573
(an mTOR inhibitor).
1298. The device of item 1192 wherein the agent is synthadotin (a tubulin antagonist). 1299. The device of item 1192 wherein the agent is S-0885 (a collagenase inhibitor).
1300. The device of item 1192 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
1301. The device of item 1192 wherein the agent is ixabepilone
(an epithilone).
1302. The device of item 1192 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1303. The device of item 1192 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1304. The device of item 1192 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1305. The device of item 1192 wherein the agent is combretastatin (an angiogenesis inhibitor).
1306. The device of item 1192 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1307. The device of item 1192 wherein the agent is SB-715992 (a kinesin antagonist).
1308. The device of item 1192 wherein the agent is temsirolimus (an mTOR inhibitor).
1309. The device of item 1192 wherein the agent is adalimumab (a TNFα antagonist).
1310. The device of item 1192, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer. 1311. The device of item 1192, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1312. The device of item 1192, further comprising a coating, wherein the coating is disposed on a surface of the device.
1313. The device of item 1192, further comprising a coating, wherein the coating directly contacts the device.
1314. The device of item 1192, further comprising a coating, wherein the coating indirectly contacts the device.
1315. The device of item 1192, further comprising a coating, wherein the coating partially covers the device.
1316. The device of item 1192, further comprising a coating, wherein the coating completely covers the device.
1317. The device of item 1192, further comprising a coating, wherein the coating is a uniform coating.
1318. The device of item 1192, further comprising a coating, wherein the coating is a non-uniform coating.
1319. The device of item 1192, further comprising a coating, wherein the coating is a discontinuous coating.
1320. The device of item 1192, further comprising a coating, wherein the coating is a patterned coating.
1321. The device of item 1192, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
1322. The device of item 1192, further comprising a coating, wherein the coating has a thickness of 10 μm or less. 1323. The device of item 1192, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1324. The device of item 1192, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1325. The device of item 1192, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1326. The device of item 1192, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1327. The device of item 1192, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1328. The device of item 1192, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1329. The device of item 1192, further comprising a coating, wherein the coating further comprises a polymer.
1330. The device of item 1192, further comprising a first coating having a first composition and the second coating having a second composition.
1331. The device of item 1192, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different. 1332. The device of item 1192, further comprising a polymer.
1333. The device of item 1192, further comprising a polymeric carrier.
1334. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1335. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1336. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1337. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1338. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1339. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1340. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1341. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1342. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1343. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer. 1344. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1345. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1346. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1347. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1348. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1349. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1350. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1351. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1352. The device of item 1192, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1353. The device of item 1192, further comprising a lubricious coating.
1354. The device of item 1192 wherein the anti-scarring agent is located within pores or holes of the device.
1355. The device of item 1192 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device. 1356. The device of item 1192, further comprising a second pharmaceutically active agent.
1357. The device of item 1192, further comprising an antiinflammatory agent.
1358. The device of item 1192, further comprising an agent that inhibits infection.
1359. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1360. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1361. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1362. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1363. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1364. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1365. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1366. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1367. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is etoposide. 1368. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1369. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1370. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1371. The device of item 1192, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1372. The device of item 1192, further comprising an anti- thrombotic agent.
1373. The device of item 1192, further comprising a visualization agent.
1374. The device of item 1192, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1375. The device of item 1192, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1376. The device of item 1192, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1377. The device of item 1192, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 1378. The device of item 1192, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1379. The device of item 1192, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1380. The device of item 1192, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1381. The device of item 1192, further comprising an echogenic material.
1382. The device of item 1192, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1383. The device of item 1192 wherein the device is sterile.
1384. The device of item 1192 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1385. The device of item 1192 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
1386. The device of item 1192 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1387. The device of item 1192 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 1388. The device of item 1192 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1389. The device of item 1192 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1390. The device of item 1192 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1391. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1392. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1393. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1394. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1395. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1396. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1397. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1398. The device of item 1192 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1399. The device of item 1192 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
1400. The device of item 1192 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
1401. The device of item 1192 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1402. The device of item 1192 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1403. The device of item 1192 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1404. The device of item 1192 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1405. The device of item 1192 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1406. The device of item 1192 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1407. The device of item 1192 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1408. The device of item 1192 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1409. The device of item 1192 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1410. The device of item 1192 wherein the agent or the composition is affixed to the sensor.
1411. The device of item 1192 wherein the agent or the composition is covalently attached to the sensor.
1412. The device of item 1192 wherein the agent or the composition is non-covalently attached to the sensor.
1413. The device of item 1192 further comprising a coating that absorbs the agent or the composition.
1414. The device of item 1192 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
1415. The device of item 1192 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
1416. The device of item 1192 wherein the sensor is completely covered with a sleeve that contains the agent or the composition. 1417. The device of item 1192 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
1418. The device of item 1192 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
1419. The device of item 1192 further comprising a pump that is linked to the sensor.
1420. The device of any one of items 1192-1419 wherein the device is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
1421. The device of any one of items 1192-1419 wherein the device generates an electrical audio signal.
1422. The device of any one of items 1192-1419 wherein the device is a capacitive sensor that is coupled to a vibrating auditory element.
1423. The device of any one of items 1192-1419 wherein the device is an electromagnetic sensor.
1424. A device, comprising an electrolyte or metabolite sensor and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1425. The device of item 1424 wherein the agent is an adensosine A2A receptor antagonist.
1426. The device of item 1424 wherein the agent is an AKT inhibitor.
1427. The device of item 1424 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects
No. 5754 (Merck KgaA). 1428. The device of item 1424 wherein the agent is an alpha 4 integrin antagonist.
1429. The device of item 1424 wherein the agent is an alpha 7 nicotinic receptor agonist.
1430. The device of item 1424 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1431. The device of item 1424 wherein the agent is an apoptosis antagonist.
1432. The device of item 1424 wherein the agent is an apoptosis activator.
1433. The device of item 1424 wherein the agent is a beta 1 integrin antagonist.
1434. The device of item 1424 wherein the agent is a beta tubulin inhibitor.
1435. The device of item 1424 wherein the agent is a blocker of enzyme production in Hepatitis C.
1436. The device of item 1424 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1437. The device of item 1424 wherein the agent is a calcineurin inhibitor.
1438. The device of item 1424 wherein the agent is a caspase 3 inhibitor.
1439. The device of item 1424 wherein the agent is a CC chemokine receptor antagonist.
1440. The device of item 1424 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 1441. The device of item 1424 wherein the agent is a cathepsin B inhibitor.
1442. The device of item 1424 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
1443. The device of item 1424 wherein the agent is a cathepsin L inhibitor.
1444. The device of item 1424 wherein the agent is a CD40 antagonist.
1445. The device of item 1424 wherein the agent is a chemokine receptor agonist.
1446. The device of item 1424 wherein the agent is a chymase inhibitor.
1447. The device of item 1424 wherein the agent is a collagenase antagonist.
1448. The device of item 1424 wherein the agent is a CXCR antagonist.
1449. The device of item 1424 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
1450. The device of item 1424 wherein the agent is a cyclooxygenase 1 inhibitor.
1451. The device of item 1424 wherein the agent is a DHFR inhibitor.
1452. The device of item 1424 wherein the agent is a dual integrin inhibitor.
1453. The device of item 1424 wherein the agent is an elastase inhibitor.
1454. The device of item 1424 wherein the agent is an elongation factor-1 alpha inhibitor.
1455. The device of item 1424 wherein the agent is an endothelial growth factor antagonist.
1456. The device of item 1424 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1457. The device of item 1424 wherein the agent is an endotoxin antagonist. 1458. The device of item 1424 wherein the agent is an epothilone and tubulin binder.
1459. The device of item 1424 wherein the agent is an estrogen receptor antagonist.
1460. The device of item 1424 wherein the agent is an FGF inhibitor.
1461. The device of item 1424 wherein the agent is a farnexyl transferase inhibitor.
1462. The device of item 1424 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1463. The device of item 1424 wherein the agent is an FLT-3 kinase inhibitor.
1464. The device of item 1424 wherein the agent is an FGF receptor kinase inhibitor.
1465. The device of item 1424 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-
13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1466. The device of item 1424 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
1467. The device of item 1424 wherein the agent is a histone deacetylase inhibitor.
1468. The device of item 1424 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
1469. The device of item 1424 wherein the agent is an ICAM inhibitor.
1470. The device of item 1424 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174
(Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1471. The device of item 1424 wherein the agent is an IL-2 inhibitor.
1472. The device of item 1424 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1473. The device of item 1424 wherein the agent is an IMPDH (inosine monophosphate).
1474. The device of item 1424 wherein the agent is an integrin antagonist.
1475. The device of item 1424 wherein the agent is an interieukin antagonist.
1476. The device of item 1424 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
1477. The device of item 1424 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1478. The device of item 1424 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1479. The device of item 1424 wherein the agent a JAK3 enzyme inhibitor.
1480. The device of item 1424 wherein the agent is a JNK inhibitor.
1481. The device of item 1424 wherein the agent is a kinase inhibitor. 1482. The device of item 1424 wherein the agent is kinesin antagonist.
1483. The device of item 1424 wherein the agent is a kinesin antagonist.
1484. The device of item 1424 wherein the agent is a ieukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1485. The device of item 1424 wherein the agent is an MAP kinase inhibitor.
1486. The device of item 1424 wherein the agent is a matrix metalloproteinase inhibitor.
1487. The device of item 1424 wherein the agent is an MCP-
CCR2 inhibitor.
1488. The device of item 1424 wherein the agent is an mTOR inhibitor. 1489. The device of item 1424 wherein the agent is an mTOR kinase inhibitor.
1490. The device of item 1424 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1491. The device of item 1424 wherein the agent is an MIF inhibitor.
1492. The device of item 1424 wherein the agent is an MMP inhibitor.
1493. The device of item 1424 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 1494. The device of item 1424 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1495. The device of item 1424 wherein the agent is a nitric oxide agonist.
1496. The device of item 1424 wherein the agent is an ornithine decarboxylase inhibitor.
1497. The device of item 1424 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1498. The device of item 1424 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
1499. The device of item 1424 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1500. The device of item 1424 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1501. The device of item 1424 wherein the agent is a phosphatase inhibitor.
1502. The device of item 1424 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1503. The device of item 1424 wherein the agent is a PKC inhibitor.
1504. The device of item 1424 wherein the agent is a platelet activating factor antagonist.
1505. The device of item 1424 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1506. The device of item 1424 wherein the agent is a prolyl hydroxylase inhibitor.
1507. The device of item 1424 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1508. The device of item 1424 wherein the agent is a protein kinase B inhibitor. 1509. The device of item 1424 wherein the agent is a protein kinase C stimulant.
1510. The device of item 1424 wherein the agent is a purine nucleoside analogue.
1511. The device of item 1424 wherein the agent is a purinoreceptor P2X antagonist.
1512. The device of item 1424 wherein the agent is a Raf kinase inhibitor.
1513. The device of item 1424 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
1514. The device of item 1424 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1515. The device of item 1424 wherein the agent is an SDF-1 antagonist.
1516. The device of item 1424 wherein the agent is a sheddase inhibitor.
1517. The device of item 1424 wherein the agent is an SRC inhibitor.
1518. The device of item 1424 wherein the agent is a stromelysin inhibitor.
1519. The device of item 1424 wherein the agent is an Syk kinase inhibitor.
1520. The device of item 1424 wherein the agent is a telomerase inhibitor. 1521. The device of item 1424 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1522. The device of item 1424 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731 -18-1 ) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajϊnomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
' 1523. The device of item 1424 wherein the agent is a Toll receptor inhibitor.
1524. The device of item 1424 wherein the agent is a tubulin antagonist.
1525. The device of item 1424 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG)1 an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 1526. The device of item 1424 wherein the agent is a VEGF inhibitor.
1527. The device of item 1424 wherein the agent is a vitamin D receptor agonist.
1528. The device of item 1424 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
1529. The device of item 1424 wherein the agent is AP-23573 (an mTOR inhibitor).
1530. The device of item 1424 wherein the agent is synthadotin (a tubulin antagonist).
1531. The device of item 1424 wherein the agent is S-0885 (a collagenase inhibitor).
1532. The device of item 1424 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
1533. The device of item 1424 wherein the agent is ixabepilone
(an epithilone).
1534. The device of item 1424 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1535. The device of item 1424 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1536. The device of item 1424 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1537. The device of item 1424 wherein the agent is combretastatin (an angiogenesis inhibitor). 1538. The device of item 1424 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1539. The device of item 1424 wherein the agent is SB-715992 (a kinesin antagonist).
1540. The device of item 1424 wherein the agent is temsirolimus (an mTOR inhibitor).
1541. The device of item 1424 wherein the agent is adalimumab (a TNFα antagonist).
1542. The device of item 1424, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
1543. The device of item 1424, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1544. The device of item 1424, further comprising a coating, wherein the coating is disposed on a surface of the device.
1545. The device of item 1424, further comprising a coating, wherein the coating directly contacts the device.
1546. The device of item 1424, further comprising a coating, wherein the coating indirectly contacts the device.
1547. The device of item 1424, further comprising a coating, wherein the coating partially covers the device.
1548. The device of item 1424, further comprising a coating, wherein the coating completely covers the device.
1549. The device of item 1424, further comprising a coating, wherein the coating is a uniform coating. 1550. The device of item 1424, further comprising a coating, wherein the coating is a non-uniform coating.
1551. The device of item 1424, further comprising a coating, wherein the coating is a discontinuous coating.
1552. The device of item 1424, further comprising a coating, wherein the coating is a patterned coating.
1553. The device of item 1424, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
1554. The device of item 1424, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
1555. The device of item 1424, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1556. The device of item 1424, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1557. The device of item 1424, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1558. The device of item 1424, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1559. The device of item 1424, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 1560. The device of item 1424, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1561. The device of item 1424, further comprising a coating, wherein the coating further comprises a polymer.
1562. The device of item 1424, further comprising a first coating having a first composition and the second coating having a second composition.
1563. The device of item 1424, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1564. The device of item 1424, further comprising a polymer.
1565. The device of item 1424, further comprising a polymeric carrier.
1566. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1567. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1568. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1569. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1570. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer. 1571. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1572. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1573. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1574. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1575. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1576. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1577. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1578. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1579. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1580. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1581. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 1582. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1583. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1584. The device of item 1424, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1585. The device of item 1424, further comprising a lubricious coating.
1586. The device of item 1424 wherein the anti-scarring agent is located within pores or holes of the device.
1587. The device of item 1424 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1588. The device of item 1424, further comprising a second pharmaceutically active agent.
1589. The device of item 1424, further comprising an antiinflammatory agent.
1590. The device of item 1424, further comprising an agent that inhibits infection.
1591. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1592. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1593. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone. 1594. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1595. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1596. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1597. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1598. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1599. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1600. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1601. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1602. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1603. The device of item 1424, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1604. The device of item 1424, further comprising an antithrombotic agent.
1605. The device of item 1424, further comprising a visualization agent. 1606. The device of item 1424, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1607. The device of item 1424, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1608. The device of item 1424, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1609. The device of item 1424, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1610. The device of item 1424, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1611. The device of item 1424, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1612. The device of item 1424, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1613. The device of item 1424, further comprising an echogenic material.
1614. The device of item 1424, further comprising an echogenic material, wherein the echogenic material is in the form of a coating. 1615. The device of item 1424 wherein the device is sterile.
1616. The device of item 1424 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1617. The device of item 1424 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
1618. The device of item 1424 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1619. The device of item 1424 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1620. The device of item 1424 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1621. The device of item 1424 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1622. The device of item 1424 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1623. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year. 1624. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1625. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1626. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1627. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1628. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1629. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1630. The device of item 1424 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1631. The device of item 1424 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
1632. The device of item 1424 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
1633. The device of item 1424 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 1634. The device of item 1424 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1635. The device of item 1424 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1636. The device of item 1424 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1637. The device of item 1424 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1638. The device of item 1424 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1639. The device of item 1424 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1640. The device of item 1424 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1641. The device of item 1424 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1642. The device of item 1424 wherein the agent or the composition is affixed to the sensor. 1643. The device of item 1424 wherein the agent or the composition is covalently attached to the sensor.
1644. The device of item 1424 wherein the agent or the composition is non-covalently attached to the sensor.
1645. The device of item 1424 further comprising a coating that absorbs the agent or the composition.
1646. The device of item 1424 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
1647. The device of item 1424 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
1648. The device of item 1424 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
1649. The device of item 1424 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
1650. The device of item 1424 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
1651. The device of item 1424 further comprising a pump that is linked to the sensor.
1652. The device of any one of items 1424-1651 wherein the device emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
1653. The device of any one of items 1424-1651 wherein the device is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader.
1654. The device of any one of items 1424-1651 wherein the device is a monolithic bioelectronic device for detecting at least one analyte within the host.
1655. A device, comprising a pump and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1656. The device of item 1655 wherein the agent is an adensosine A2A receptor antagonist.
1657. The device of item 1655 wherein the agent is an AKT inhibitor.
1658. The device of item 1655 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
1659. The device of item 1655 wherein the agent is an alpha 4 integrin antagonist.
1660. The device of item 1655 wherein the agent is an alpha 7 nicotinic receptor agonist.
1661. The device of item 1655 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH)1 ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005
(GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1662. The device of item 1655 wherein the agent is an apoptosis antagonist.
1663. The device of item 1655 wherein the agent is an apoptosis activator.
1664. The device of item 1655 wherein the agent is a beta 1 integrin antagonist. 1665. The device of item 1655 wherein the agent is a beta tubulin inhibitor.
1666. The device of item 1655 wherein the agent is a blocker of enzyme production in Hepatitis C.
1667. The device of item 1655 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1668. The device of item 1655 wherein the agent is a calcineurin inhibitor.
1669. The device of item 1655 wherein the agent is a caspase 3 inhibitor.
1670. The device of item 1655 wherein the agent is a CC chemokine receptor antagonist.
1671. The device of item 1655 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
1672. The device of item 1655 wherein the agent is a cathepsin B inhibitor.
1673. The device of item 1655 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
1674. The device of item 1655 wherein the agent is a cathepsin L inhibitor.
1675. The device of item 1655 wherein the agent is a CD40 antagonist. 1676. The device of item 1655 wherein the agent is a chemokine receptor agonist.
1677. The device of item 1655 wherein the agent is a chymase inhibitor.
1678. The device of item 1655 wherein the agent is a collagenase antagonist.
1679. The device of item 1655 wherein the agent is a CXCR antagonist.
1680. The device of item 1655 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
1681. The device of item 1655 wherein the agent is a cyclooxygenase 1 inhibitor.
1682. The device of item 1655 wherein the agent is a DHFR inhibitor.
1683. The device of item 1655 wherein the agent is a dual integrin inhibitor.
1684. The device of item 1655 wherein the agent is an elastase inhibitor. 1685. The device of item 1655 wherein the agent is an elongation factor-1 alpha inhibitor.
1686. The device of item 1655 wherein the agent is an endothelial growth factor antagonist.
1687. The device of item 1655 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1688. The device of item 1655 wherein the agent is an endotoxin antagonist.
1689. The device of item 1655 wherein the agent is an epothilone and tubulin binder.
1690. The device of item 1655 wherein the agent is an estrogen receptor antagonist.
1691. The device of item 1655 wherein the agent is an FGF inhibitor.
1692. The device of item 1655 wherein the agent is a famexyl transferase inhibitor.
1693. The device of item 1655 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Sen/ier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1694. The device of item 1655 wherein the agent is an FLT-3 kinase inhibitor.
1695. The device of item 1655 wherein the agent is an FGF receptor kinase inhibitor.
1696. The device of item 1655 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-
13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1697. The device of item 1655 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
1698. The device of item 1655 wherein the agent is a histone deacetylase inhibitor.
1699. The device of item 1655 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 1700. The device of item 1655 wherein the agent is an ICAM inhibitor.
1701. The device of item 1655 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1702. The device of item 1655 wherein the agent is an IL-2 inhibitor.
1703. The device of item 1655 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1704. The device of item 1655 wherein the agent is an IMPDH (inosine monophosphate). 1705. The device of item 1655 wherein the agent is an integrin antagonist.
1706. The device of item 1655 wherein the agent is an interleukin antagonist.
1707. The device of item 1655 wherein the agent is an inhibitor of type 111 receptor tyrosine kinase.
1708. The device of item 1655 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1709. The device of item 1655 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1710. The device of item 1655 wherein the agent a JAK3 enzyme inhibitor.
1711. The device of item 1655 wherein the agent is a JNK inhibitor.
1712. The device of item 1655 wherein the agent is a kinase inhibitor.
1713. The device of item 1655 wherein the agent is kinesin antagonist.
1714. The device of item 1655 wherein the agent is a kinesin antagonist.
1715. The device of item 1655 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1716. The device of item 1655 wherein the agent is an MAP kinase inhibitor.
1717. The device of item 1655 wherein the agent is a matrix metalloproteinase inhibitor.
1718. The device of item 1655 wherein the agent is an MCP-
CCR2 inhibitor.
1719. The device of item 1655 wherein the agent is an mTOR inhibitor.
1720. The device of item 1655 wherein the agent is an mTOR kinase inhibitor.
1721. The device of item 1655 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1722. The device of item 1655 wherein the agent is an MIF inhibitor.
1723. The device of item 1655 wherein the agent is an MMP inhibitor.
1724. The device of item 1655 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1725. The device of item 1655 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 1726. The device of item 1655 wherein the agent is a nitric oxide agonist.
1727. The device of item 1655 wherein the agent is an ornithine decarboxylase inhibitor.
1728. The device of item 1655 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1729. The device of item 1655 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
1730. The device of item 1655 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1731. The device of item 1655 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1732. The device of item 1655 wherein the agent is a phosphatase inhibitor.
1733. The device of item 1655 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosai, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1734. The device of item 1655 wherein the agent is a PKC inhibitor.
1735. The device of item 1655 wherein the agent is a platelet activating factor antagonist.
1736. The device of item 1655 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1737. The device of item 1655 wherein the agent is a prolyl hydroxylase inhibitor.
1738. The device of item 1655 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1739. The device of item 1655 wherein the agent is a protein kinase B inhibitor.
1740. The device of item 1655 wherein the agent is a protein kinase C stimulant.
1741. The device of item 1655 wherein the agent is a purine nucleoside analogue.
1742. The device of item 1655 wherein the agent is a purinoreceptor P2X antagonist.
1743. The device of item 1655 wherein the agent is a Raf kinase inhibitor. 1744. The device of item 1655 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
1745. The device of item 1655 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1746. The device of item 1655 wherein the agent is an SDF-1 antagonist.
1747. The device of item 1655 wherein the agent is a sheddase inhibitor.
1748. The device of item 1655 wherein the agent is an SRC inhibitor.
1749. The device of item 1655 wherein the agent is a stromelysin inhibitor.
1750. The device of item 1655 wherein the agent is an Syk kinase inhibitor.
1751. The device of item 1655 wherein the agent is a telomerase inhibitor.
1752. The device of item 1655 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme
(Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1753. The device of item 1655 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1754. The device of item 1655 wherein the agent is a Toll receptor inhibitor.
1755. The device of item 1655 wherein the agent is a tubulin antagonist.
1756. The device of item 1655 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmith Kline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1757. The device of item 1655 wherein the agent is a VEGF inhibitor.
1758. The device of item 1655 wherein the agent is a vitamin D receptor agonist.
1759. The device of item 1655 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
1760. The device of item 1655 wherein the agent is AP-23573 (an mTOR inhibitor). 1761. The device of item 1655 wherein the agent is synthadotin (a tubulin antagonist).
1762. The device of item 1655 wherein the agent is S-0885 (a collagenase inhibitor).
1763. The device of item 1655 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
1764. The device of item 1655 wherein the agent is ixabepilone (an epithilone).
1765. The device of item 1655 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1766. The device of item 1655 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1767. The device of item 1655 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1768. The device of item 1655 wherein the agent is combretastatin (an angiogenesis inhibitor).
1769. The device of item 1655 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1770. The device of item 1655 wherein the agent is SB-715992 (a kinesin antagonist).
1771. The device of item 1655 wherein the agent is temsirolimus (an mTOR inhibitor).
1772. The device of item 1655 wherein the agent is adalimumab (a TNFα antagonist). 1773. The device of item 1655, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
1774. The device of item 1655, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1775. The device of item 1655, further comprising a coating, wherein the coating is disposed on a surface of the device.
1776. The device of item 1655, further comprising a coating, wherein the coating directly contacts the device.
1777. The device of item 1655, further comprising a coating, wherein the coating indirectly contacts the device.
1778. The device of item 1655, further comprising a coating, wherein the coating partially covers the device.
1779. The device of item 1655, further comprising a coating, wherein the coating completely covers the device.
1780. The device of item 1655, further comprising a coating, wherein the coating is a uniform coating.
1781. The device of item 1655, further comprising a coating, wherein the coating is a non-uniform coating.
1782. The device of item 1655, further comprising a coating, wherein the coating is a discontinuous coating.
1783. The device of item 1655, further comprising a coating, wherein the coating is a patterned coating.
1784. The device of item 1655, further comprising a coating, wherein the coating has a thickness of 100 μm or less. 1785. The device of item 1655, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
1786. The device of item 1655, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1787. The device of item 1655, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1788. The device of item 1655, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
1789. The device of item 1655, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1790. The device of item 1655, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1791. The device of item 1655, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1792. The device of item 1655, further comprising a coating, wherein the coating further comprises a polymer.
1793. The device of item 1655, further comprising a first coating having a first composition and the second coating having a second composition.
1794. The device of item 1655, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1795. The device of item 1655, further comprising a polymer.
1796. The device of item 1655, further comprising a polymeric carrier.
1797. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1798. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1799. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1800. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1801. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1802. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1803. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1804. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1805. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains. 1806. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1807. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1808. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1809. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1810. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1811. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1812. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1813. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1814. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1815. The device of item 1655, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1816. The device of item 1655, further comprising a lubricious coating.
1817. The device of item 1655 wherein the anti-scarring agent is located within pores or holes of the device. 1818. The device of item 1655 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1819. The device of item 1655, further comprising a second pharmaceutically active agent.
1820. The device of item 1655, further comprising an antiinflammatory agent.
1821. The device of item 1655, further comprising an agent that inhibits infection.
1822. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1823. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1824. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1825. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1826. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1827. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1828. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1829. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin. 1830. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1831. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1832. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1833. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1834. The device of item 1655, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1835. The device of item 1655, further comprising an antithrombotic agent.
1836. The device of item 1655, further comprising a visualization agent.
1837. The device of item 1655, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1838. The device of item 1655, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1839. The device of item 1655, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material. 1840. The device of item 1655, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1841. The device of item 1655, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1842. The device of item 1655, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1843. The device of item 1655, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1844. The device of item 1655, further comprising an echogenic material.
1845. The device of item 1655, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1846. The device of item 1655 wherein the device is sterile.
1847. The device of item 1655 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1848. The device of item 1655 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
1849. The device of item 1655 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. 1850. The device of item 1655 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1851. The device of item 1655 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1852. The device of item 1655 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1853. The device of item 1655 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1854. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1855. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1856. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1857. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1858. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 1859. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1860. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1861. The device of item 1655 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1862. The device of item 1655 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
1863. The device of item 1655 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
1864. The device of item 1655 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1865. The device of item 1655 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1866. The device of item 1655 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1867. The device of item 1655 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1868. The device of item 1655 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1869. The device of item 1655 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1870. The device of item 1655 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1871. The device of item 1655 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1872. The device of item 1655 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1873. The device of item 1655 wherein the agent or the composition is affixed to the sensor.
1874. The device of item 1655 wherein the agent or the composition is covalently attached to the sensor.
1875. The device of item 1655 wherein the agent or the composition is non-covalently attached to the sensor.
1876. The device of item 1655 further comprising a coating that absorbs the agent or the composition.
1877. The device of item 1655 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
1878. The device of item 1655 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition. 1879. The device of item 1655 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
1880. The device of item 1655 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
1881. The device of item 1655 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
1882. The device of item 1655 further comprising a pump that is linked to the sensor.
1883. The device of any one of items 1655-1882 wherein the device is adapted for delivering insulin.
1884. The device of any one of items 1655-1882 wherein the device is adapted for delivering a narcotic.
1885. The device of any one of items 1655-1882 wherein the device is adapted for delivering a chemotherapeutic agent.
1886. The device of any one of items 1655-1882 wherein the device is adapted for delivering an anti-arrhythmic drug.
1887. The device of any one of items 1655-1882 wherein the device is adapted for delivering an anti-spasmotic drug.
1888. The device of any one of items 1655-1882 wherein the device is adapted for delivering an anti-spastic agent.
1889. The device of any one of items 1655-1882 wherein the device is adapted for delivering an antibiotic.
1890. The device of any one of items 1655-1882 wherein the device is adapted for delivering a drug only when changes in the host are detected. 1891. The device of any one of items 1655-1882 wherein the device is adapted for delivering a drug as a continuous slow release.
1892. The device of any one of items 1655-1882 wherein the device is adapted for delivering a drug at prescribed dosages in a pulsatile manner.
1893. The device of any one of items 1655-1882 wherein the device is a programmable drug delivery pump.
1894. The device of any one of items 1655-1882 wherein the device is adapted for intraocularly delivering a drug.
1895. The device of any one of items 1655-1882 wherein the device is adapted for intrathecal^ delivering a drug.
1896. The device of any one of items 1655-1882 wherein the device is adapted for intraperitoneally delivering a drug.
1897. The device of any one of items 1655-1882 wherein the device is adapted for intra-arterially delivering a drug.
1898. The device of any one of items 1655-1882 wherein the device is adapted for intracardiac delivery of a drug.
1899. The device of any one of items 1655-1882 wherein the device is an implantable osmotic pump.
1900. The device of any one of items 1655-1882 wherein the device is an ocular drug delivery pump.
1901. The device of any one of items 1655-1882 wherein the device is metering system.
1902. The device of any one of items 1655-1882 wherein the device is a peristaltic (roller) pump. 1903. The device of any one of items 1655-1882 wherein the device is an electronically driven pump.
1904. The device of any one of items 1655-1882 wherein the device is an elastomeric pump.
1905. The device of any one of items 1655-1882 wherein the device is a spring contraction pump.
1906. The device of any one of items 1655-1882 wherein the device is a gas-driven pump.
1907. The device of any one of items 1655-1882 wherein the device is a hydraulic pump.
1908. The device of any one of items 1655-1882 wherein the device is a piston-dependent pump.
1909. The device of any one of items 1655-1882 wherein the device is a non-piston-dependent pump.
1910. The device of any one of items 1655-1882 wherein the device is a dispensing chamber.
1911. The device of any one of items 1655-1882 wherein the device is an infusion pump.
1912. The device of any one of items 1655-1882 wherein the device is a passive pump.
1913. A device, comprising an implantable insulin pump and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 1914. The device of item 1913 wherein the agent is an adensosine A2A receptor antagonist.
1915. The device of item 1913 wherein the agent is an AKT inhibitor.
1916. The device of item 1913 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
1917. The device of item 1913 wherein the agent is an alpha 4 integrin antagonist.
1918. The device of item 1913 wherein the agent is an alpha 7 nicotinic receptor agonist.
1919. The device of item 1913 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant
Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-
23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1920. The device of item 1913 wherein the agent is an apoptosis antagonist.
1921. The device of item 1913 wherein the agent is an apoptosis activator.
1922. The device of item 1913 wherein the agent is a beta 1 integrin antagonist.
1923. The device of item 1913 wherein the agent is a beta tubulin inhibitor.
1924. The device of item 1913 wherein the agent is a blocker of enzyme production in Hepatitis C.
1925. The device of item 1913 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1926. The device of item 1913 wherein the agent is a calcineurin inhibitor. 1927. The device of item 1913 wherein the agent is a caspase 3 inhibitor.
1928. The device of item 1913 wherein the agent is a CC chemokine receptor antagonist.
1929. The device of item 1913 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
1930. The device of item 1913 wherein the agent is a cathepsin B inhibitor.
1931. The device of item 1913 wherein the agent is a cathepsin
K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
1932. The device of item 1913 wherein the agent is a cathepsin L inhibitor.
1933. The device of item 1913 wherein the agent is a CD40 antagonist.
1934. The device of item 1913 wherein the agent is a chemokine receptor agonist.
1935. The device of item 1913 wherein the agent is a chymase inhibitor.
1936. The device of item 1913 wherein the agent is a collagenase antagonist.
1937. The device of item 1913 wherein the agent is a CXCR antagonist. 1938. The device of item 1913 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
1939. The device of item 1913 wherein the agent is a cyclooxygenase 1 inhibitor.
1940. The device of item 1913 wherein the agent is a DHFR inhibitor.
1941. The device of item 1913 wherein the agent is a dual integrin inhibitor.
1942. The device of item 1913 wherein the agent is an elastase inhibitor.
1943. The device of item 1913 wherein the agent is an elongation factor-1 alpha inhibitor.
1944. The device of item 1913 wherein the agent is an endothelial growth factor antagonist.
1945. The device of item 1913 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1946. The device of item 1913 wherein the agent is an endotoxin antagonist.
1947. The device of item 1913 wherein the agent is an epothilone and tubulin binder.
1948. The device of item 1913 wherein the agent Ts an estrogen receptor antagonist.
1949. The device of item 1913 wherein the agent is an FGF inhibitor.
1950. The device of item 1913 wherein the agent is a farnexyl transferase inhibitor.
1951. The device of item 1913 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1952. The device of item 1913 wherein the agent is an FLT-3 kinase inhibitor.
1953. The device of item 1913 wherein the agent is an FGF receptor kinase inhibitor. 1954. The device of item 1913 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1955. The device of item 1913 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4~didehydro-1-deoxy-1 ,4~dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
1956. The device of item 1913 wherein the agent is a histone deacetylase inhibitor.
1957. The device of item 1913 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
1958. The device of item 1913 wherein the agent is an ICAM inhibitor.
1959. The device of item 1913 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1960. The device of item 1913 wherein the agent is an IL-2 inhibitor. 1961. The device of item 1913 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1962. The device of item 1913 wherein the agent is an IMPDH (inosine monophosphate).
1963. The device of item 1913 wherein the agent is an integrin antagonist.
1964. The device of item 1913 wherein the agent is an interleukin antagonist.
1965. The device of item 1913 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
1966. The device of item 1913 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 1967. The device of item 1913 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1968. The device of item 1913 wherein the agent a JAK3 enzyme inhibitor.
1969. The device of item 1913 wherein the agent is a JNK inhibitor.
1970. The device of item 1913 wherein the agent is a kinase inhibitor.
1971. The device of item 1913 wherein the agent is kinesin antagonist.
1972. The device of item 1913 wherein the agent is a kinesin antagonist.
1973. The device of item 1913 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No.
346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof. 1974. The device of item 1913 wherein the agent is an MAP kinase inhibitor.
1975. The device of item 1913 wherein the agent is a matrix metalloproteinase inhibitor.
1976. The device of item 1913 wherein the agent is an MCP-
CCR2 inhibitor.
1977. The device of item 1913 wherein the agent is an mTOR inhibitor.
1978. The device of item 1913 wherein the agent is an mTOR kinase inhibitor.
1979. The device of item 1913 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1980. The device of item 1913 wherein the agent is an MIF inhibitor.
1981. The device of item 1913 wherein the agent is an MMP inhibitor. 1982. The device of item 1913 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1983. The device of item 1913 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-
576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1984. The device of item 1913 wherein the agent is a nitric oxide agonist.
1985. The device of item 1913 wherein the agent is an ornithine decarboxylase inhibitor.
1986. The device of item 1913 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1987. The device of item 1913 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
1988. The device of item 1913 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1989. The device of item 1913 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1990. The device of item 1913 wherein the agent is a phosphatase inhibitor.
1991. The device of item 1913 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1992. The device of item 1913 wherein the agent is a PKC inhibitor.
1993. The device of item 1913 wherein the agent is a platelet activating factor antagonist. 1994. The device of item 1913 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1995. The device of item 1913 wherein the agent is a prolyl hydroxylase inhibitor.
1996. The device of item 1913 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1997. The device of item 1913 wherein the agent is a protein kinase B inhibitor.
1998. The device of item 1913 wherein the agent is a protein kinase C stimulant.
1999. The device of item 1913 wherein the agent is a purine nucleoside analogue.
2000. The device of item 1913 wherein the agent is a purinoreceptor P2X antagonist.
2001. The device of item 1913 wherein the agent is a Raf kinase inhibitor.
2002. The device of item 1913 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2003. The device of item 1913 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2004. The device of item 1913 wherein the agent is an SDF-1 antagonist.
2005. The device of item 1913 wherein the agent is a sheddase inhibitor. 2006. The device of item 1913 wherein the agent is an SRC inhibitor.
2007. The device of item 1913 wherein the agent is a stromelysin inhibitor.
2008. The device of item 1913 wherein the agent is an Syk kinase inhibitor.
2009. The device of item 1913 wherein the agent is a telomerase inhibitor.
2010. The device of item 1913 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-
8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
2011. The device of item 1913 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2012. The device of item 1913 wherein the agent is a Toll receptor inhibitor.
2013. The device of item 1913 wherein the agent is a tubulin antagonist. 2014. The device of item 1913 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or t GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2015. The device of item 1913 wherein the agent is a VEGF inhibitor.
2016. The device of item 1913 wherein the agent is a vitamin D receptor agonist.
2017. The device of item 1913 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
2018. The device of item 1913 wherein the agent is AP-23573 (an mTOR inhibitor).
2019. The device of item 1913 wherein the agent is synthadotin (a tubulin antagonist).
2020. The device of item 1913 wherein the agent is S-0885 (a collagenase inhibitor).
2021. The device of item 1913 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2022. The device of item 1913 wherein the agent is ixabepilone
(an epithilone). 2023. The device of item 1913 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2024. The device of item 1913 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2025. The device of item 1913 wherein the agent is ABT-518
(an angiogenesis inhibitor).
2026. The device of item 1913 wherein the agent is combretastatin (an angiogenesis inhibitor).
2027. The device of item 1913 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2028. The device of item 1913 wherein the agent is SB-715992 (a kinesin antagonist).
2029. The device of item 1913 wherein the agent is temsirolimus (an mTOR inhibitor).
2030. The device of item 1913 wherein the agent is adalimumab (a TNFα antagonist).
2031. The device of item 1913, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
2032. The device of item 1913, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2033. The device of item 1913, further comprising a coating, wherein the coating is disposed on a surface of the device.
2034. The device of item 1913, further comprising a coating, wherein the coating directly contacts the device. 2035. The device of item 1913, further comprising a coating, wherein the coating indirectly contacts the device.
2036. The device of item 1913, further comprising a coating, wherein the coating partially covers the device.
2037. The device of item 1913, further comprising a coating, wherein the coating completely covers the device.
2038. The device of item 1913, further comprising a coating, wherein the coating is a uniform coating.
2039. The device of item 1913, further comprising a coating, wherein the coating is a non-uniform coating.
2040. The device of item 1913, further comprising a coating, wherein the coating is a discontinuous coating.
2041. The device of item 1913, further comprising a coating, wherein the coating is a patterned coating.
2042. The device of item 1913, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
2043. The device of item 1913, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
2044. The device of item 1913, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2045. The device of item 1913, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year. 2046. The device of item 1913, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2047. The device of item 1913, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
2048. The device of item 1913, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2049. The device of item 1913, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2050. The device of item 1913, further comprising a coating, wherein the coating further comprises a polymer.
2051. The device of item 1913, further comprising a first coating having a first composition and the second coating having a second composition.
2052. The device of item 1913, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2053. The device of item 1913, further comprising a polymer.
2054. The device of item 1913, further comprising a polymeric carrier.
2055. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer. 2056. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2057. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2058. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2059. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2060. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2061. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2062. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2063. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2064. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2065. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2066. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel. 2067. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2068. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2069. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2070. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2071. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2072. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2073. The device of item 1913, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2074. The device of item 1913, further comprising a lubricious coating.
2075. The device of item 1913 wherein the anti-scarring agent is located within pores or holes of the device.
2076. The device of item 1913 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2077. The device of item 1913, further comprising a second pharmaceutically active agent.
2078. The device of item 1913, further comprising an anti- inflammatory agent. 2079. The device of item 1913, further comprising an agent that inhibits infection.
2080. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2081. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2082. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2083. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2084. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2085. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2086. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2087. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2088. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2089. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2090. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 2091. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2092. The device of item 1913, further comprising an agent that inhibits infection, wherein the agent is cispiatin.
2093. The device of item 1913, further comprising an antithrombotic agent.
2094. The device of item 1913, further comprising a visualization agent.
2095. The device of item 1913, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2096. The device of item 1913, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2097. The device of item 1913, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2098. The device of item 1913, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2099. The device of item 1913, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 2100. The device of item 1913, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2101. The device of item 1913, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2102. The device of item 1913, further comprising an echogenic material.
2103. The device of item 1913, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2104. The device of item 1913 wherein the device is sterile.
2105. The device of item 1913 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2106. The device of item 1913 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
2107. The device of item 1913 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2108. The device of item 1913 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2109. The device of item 1913 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue. 2110. The device of item 1913 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2111. The device of item 1913 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2112. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2113. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2114. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2115. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2116. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2117. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2118. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 2119. The device of item 1913 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2120. The device of item 1913 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
2121. The device of item 1913 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
2122. The device of item 1913 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2123. The device of item 1913 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2124. The device of item 1913 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2125. The device of item 1913 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2126. The device of item 1913 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2127. The device of item 1913 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2128. The device of item 1913 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2129. The device of item 1913 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2130. The device of item 1913 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2131. The device of item 1913 wherein the agent or the composition is affixed to the sensor.
2132. The device of item 1913 wherein the agent or the composition is covalently attached to the sensor.
2133. The device of item 1913 wherein the agent or the composition is non-covalently attached to the sensor.
2134. The device of item 1913 further comprising a coating that absorbs the agent or the composition.
2135. The device of item 1913 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
2136. The device of item 1913 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
2137. The device of item 1913 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
2138. The device of item 1913 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
2139. The device of item 1913 wherein the sensor is completely covered with a mesh that contains the agent or the composition. 2140. The device of item 1913 further comprising a pump that is linked to the sensor.
2141. The device of any one of items 1913-2140 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense mediation through the catheter.
2142. A device, comprising an intrathecal drug delivery pump and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2143. The device of item 2142 wherein the agent is an adensosine A2A receptor antagonist.
2144. The device of item 2142 wherein the agent is an AKT inhibitor.
2145. The device of item 2142 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
2146. The device of item 2142 wherein the agent is an alpha 4 integrin antagonist.
2147. The device of item 2142 wherein the agent is an alpha 7 nicotinic receptor agonist.
2148. The device of item 2142 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Ceigene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Ceigene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Ceigene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPl-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2149. The device of item 2142 wherein the agent is an apoptosis antagonist.
2150. The device of item 2142 wherein the agent is an apoptosis activator. 2151. The device of item 2142 wherein the agent is a beta 1 integrin antagonist.
2152. The device of item 2142 wherein the agent is a beta tubulin inhibitor.
2153. The device of item 2142 wherein the agent is a blocker of enzyme production in Hepatitis C.
2154. The device of item 2142 wherein the agent is a Bruton's tyrosine kinase inhibitor.
2155. The device of item 2142 wherein the agent is a calcineurin inhibitor.
2156. The device of item 2142 wherein the agent is a caspase 3 inhibitor.
2157. The device of item 2142 wherein the agent is a CC chemokine receptor antagonist.
2158. The device of item 2142 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2159. The device of item 2142 wherein the agent is a cathepsin B inhibitor.
2160. The device of item 2142 wherein the agent is a cathepsin
K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
2161. The device of item 2142 wherein the agent is a cathepsin L inhibitor. 2162. The device of item 2142 wherein the agent is a CD40 antagonist.
2163. The device of item 2142 wherein the agent is a chemokine receptor agonist.
2164. The device of item 2142 wherein the agent is a chymase inhibitor.
2165. The device of item 2142 wherein the agent is a coliagenase antagonist.
2166. The device of item 2142 wherein the agent is a CXCR antagonist.
2167. The device of item 2142 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2168. The device of item 2142 wherein the agent is a cyclooxygenase 1 inhibitor.
2169. The device of item 2142 wherein the agent is a DHFR inhibitor.
2170. The device of item 2142 wherein the agent is a dual integrin inhibitor. 2171. The device of item 2142 wherein the agent is an elastase inhibitor.
2172. The device of item 2142 wherein the agent is an elongation factor-1 alpha inhibitor.
2173. The device of item 2142 wherein the agent is an endothelial growth factor antagonist.
2174. The device of item 2142 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2175. The device of item 2142 wherein the agent is an endotoxin antagonist.
2176. The device of item 2142 wherein the agent is an epothilone and tubulin binder.
2177. The device of item 2142 wherein the agent is an estrogen receptor antagonist.
2178. The device of item 2142 wherein the agent is an FGF inhibitor.
2179. The device of item 2142 wherein the agent is a farnexyl transferase inhibitor. 2180. The device of item 2142 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2181. The device of item 2142 wherein the agent is an FLT-3 kinase inhibitor.
2182. The device of item 2142 wherein the agent is an FGF receptor kinase inhibitor.
2183. The device of item 2142 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
2184. The device of item 2142 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
2185. The device of item 2142 wherein the agent is a histone deacetylase inhibitor.
2186. The device of item 2142 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
2187. The device of item 2142 wherein the agent is an ICAM inhibitor.
2188. The device of item 2142 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2189. The device of item 2142 wherein the agent is an IL-2 inhibitor.
2190. The device of item 2142 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 2191. The device of item 2142 wherein the agent is an IMPDH (inosine monophosphate).
2192. The device of item 2142 wherein the agent is an integrin antagonist.
2193. The device of item 2142 wherein the agent is an interleukin antagonist.
2194. The device of item 2142 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
2195. The device of item 2142 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
2196. The device of item 2142 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
2197. The device of item 2142 wherein the agent a JAK3 enzyme inhibitor.
2198. The device of item 2142 wherein the agent is a JNK inhibitor.
2199. The device of item 2142 wherein the agent is a kinase inhibitor.
2200. The device of item 2142 wherein the agent is kinesin antagonist.
2201. The device of item 2142 wherein the agent is a kinesin antagonist.
2202. The device of item 2142 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2203. The device of item 2142 wherein the agent is an MAP kinase inhibitor.
2204. The device of item 2142 wherein the agent is a matrix metalloproteinase inhibitor.
2205. The device of item 2142 wherein the agent is an MCP- CCR2 inhibitor.
2206. The device of item 2142 wherein the agent is an mTOR inhibitor.
2207. The device of item 2142 wherein the agent is an mTOR kinase inhibitor.
2208. The device of item 2142 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
2209. The device of item 2142 wherein the agent is an MIF inhibitor.
2210. The device of item 2142 wherein the agent is an MMP inhibitor.
2211. The device of item 2142 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2212. The device of item 2142 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis); bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
2213. The device of item 2142 wherein the agent is a nitric oxide agonist.
2214. The device of item 2142 wherein the agent is an ornithine decarboxylase inhibitor.
2215. The device of item 2142 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
2216. The device of item 2142 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
2217. The device of item 2142 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSl Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2218. The device of item 2142 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biqsciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2219. The device of item 2142 wherein the agent is a phosphatase inhibitor.
2220. The device of item 2142 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
2221. The device of item 2142 wherein the agent is a PKC inhibitor.
2222. The device of item 2142 wherein the agent is a platelet activating factor antagonist.
2223. The device of item 2142 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2224. The device of item 2142 wherein the agent is a prolyl hydroxylase inhibitor.
2225. The device of item 2142 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2226. The device of item 2142 wherein the agent is a protein kinase B inhibitor.
2227. The device of item 2142 wherein the agent is a protein kinase C stimulant.
2228. The device of item 2142 wherein the agent is a purine nucleoside analogue.
2229. The device of item 2142 wherein the agent is a purinoreceptor P2X antagonist. 2230. The device of item 2142 wherein the agent is a Raf kinase inhibitor.
2231. The device of item 2142 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2232. The device of item 2142 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2233. The device of item 2142 wherein the agent is an SDF-1 antagonist.
2234. The device of item 2142 wherein the agent is a sheddase inhibitor.
2235. The device of item 2142 wherein the agent is an SRC inhibitor.
2236. The device of item 2142 wherein the agent is a stromelysin inhibitor.
2237. The device of item 2142 wherein the agent is an Syk kinase inhibitor.
2238. The device of item 2142 wherein the agent is a telomerase inhibitor.
2239. The device of item 2142 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-
8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 2240. The device of item 2142 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck. & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2241. The device of item 2142 wherein the agent is a Toll receptor inhibitor.
2242. The device of item 2142 wherein the agent is a tubulin antagonist.
2243. The device of item 2142 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2244. The device of item 2142 wherein the agent is a VEGF inhibitor.
2245. The device of item 2142 wherein the agent is a vitamin D receptor agonist.
2246. The device of item 2142 wherein the agent is ZD-6474 (an angiogenesis inhibitor). 2247. The device of item 2142 wherein the agent is AP-23573 (an mTOR inhibitor).
2248. The device of item 2142 wherein the agent is synthadotin (a tubulin antagonist).
2249. The device of item 2142 wherein the agent is S-0885 (a collagenase inhibitor).
2250. The device of item 2142 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2251. The device of item 2142 wherein the agent is ixabepilone (an epithilone).
2252. The device of item 2142 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2253. The device of item 2142 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2254. The device of item 2142 wherein the agent is ABT-518
(an angiogenesis inhibitor).
2255. The device of item 2142 wherein the agent is combretastatin (an angiogenesis inhibitor).
2256. The device of item 2142 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2257. The device of item 2142 wherein the agent is SB-715992 (a kinesin antagonist).
2258. The device of item 2142 wherein the agent is temsirolimus (an mTOR inhibitor). 2259. The device of item 2142 wherein the agent is adalimumab (a TNFα antagonist).
2260. The device of item 2142, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
2261. The device of item 2142, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2262. The device of item 2142, further comprising a coating, wherein the coating is disposed on a surface of the device.
2263. The device of item 2142, further comprising a coating, wherein the coating directly contacts the device.
2264. The device of item 2142, further comprising a coating, wherein the coating indirectly contacts the device.
2265. The device of item 2142, further comprising a coating, wherein the coating partially covers the device.
2266. The device of item 2142, further comprising a coating, wherein the coating completely covers the device.
2267. The device of item 2142, further comprising a coating, wherein the coating is a uniform coating.
2268. The device of item 2142, further comprising a coating, wherein the coating is a non-uniform coating.
2269. The device of item 2142, further comprising a coating, wherein the coating is a discontinuous coating.
2270. The device of item 2142, further comprising a coating, wherein the coating is a patterned coating. 2271. The device of item 2142, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
2272. The device of item 2142, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
2273. The device of item 2142, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2274. The device of item 2142, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2275. The device of item 2142, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2276. The device of item 2142, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
2277. The device of item 2142, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2278. The device of item 2142, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2279. The device of item 2142, further comprising a coating, wherein the coating further comprises a polymer.
2280. The device of item 2142, further comprising a first coating having a first composition and the second coating having a second composition. 2281. The device of item 2142, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2282. The device of item 2142, further comprising a polymer.
2283. The device of item 2142, further comprising a polymeric carrier.
2284. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2285. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2286. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2287. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2288. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2289. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2290. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2291. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains. 2292. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2293. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2294. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2295. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2296. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2297. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2298. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2299. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2300. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2301. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2302. The device of item 2142, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer. 2303. The device of item 2142, further comprising a lubricious coating.
2304. The device of item 2142 wherein the anti-scarring agent is located within pores or holes of the device.
2305. The device of item 2142 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2306. The device of item 2142, further comprising a second pharmaceutically active agent.
2307. The device of item 2142, further comprising an anti- inflammatory agent.
2308. The device of item 2142, further comprising an agent that inhibits infection.
2309. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2310. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2311. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2312. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2313. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2314. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. 2315. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2316. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2317. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2318. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2319. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2320. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2321. The device of item 2142, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2322. The device of item 2142, further comprising an antithrombotic agent.
2323. The device of item 2142, further comprising a visualization agent.
2324. The device of item 2142, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2325. The device of item 2142, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium. 2326. The device of item 2142, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2327. The device of item 2142, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2328. The device of item 2142, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2329. The device of item 2142, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2330. The device of item 2142, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2331. The device of item 2142, further comprising an echogenic material.
2332. The device of item 2142, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2333. The device of item 2142 wherein the device is sterile.
2334. The device of item 2142 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2335. The device of item 2142 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device. 2336. The device of item 2142 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2337. The device of item 2142 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2338. The device of item 2142 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2339. The device of item 2142 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2340. The device of item 2142 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2341. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2342. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2343. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2344. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate. 2345. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2346. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2347. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2348. The device of item 2142 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2349. The device of item 2142 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
2350. The device of item 2142 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
2351. The device of item 2142 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2352. The device of item 2142 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2353. The device of item 2142 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2354. The device of item 2142 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2355. The device of item 2142 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2356. The device of item 2142 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2357. The device of item 2142 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2358. The device of item 2142 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2359. The device of item 2142 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2360. The device of item 2142 wherein the agent or the composition is affixed to the sensor.
2361. The device of item 2142 wherein the agent or the composition is covalently attached to the sensor.
2362. The device of item 2142 wherein the agent or the composition is non-covalently attached to the sensor.
2363. The device of item 2142 further comprising a coating that absorbs the agent or the composition. 2364. The device of item 2142 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
2365. The device of item 2142 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
2366. The device of item 2142 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
2367. The device of item 2142 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
2368. The device of item 2142 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
2369. The device of item 2142 further comprising a pump that is linked to the sensor.
2370. The device of any one of items 2142-2369 wherein the device is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord.
2371. The device of any one of items 2142-2369 wherein the device is adapted for delivering a drug to the brain.
2372. The device of any one of items 2142-2369 wherein the device is adapted for intrathecal delivering baclofen.
2373. The device of any one of items 2142-2369 wherein the > device further comprises an intraspinal catheter.
2374. The device of any one of items 2142-2369 further comprising a second intrathecal drug delivery pump. 2375. The device of any one of items 2142-2369 wherein the device further comprises a catheter and an electrode.
2376. A device, comprising an implantable drug delivery pump for chemotherapy and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2377. The device of item 2376 wherein the agent is an adensosine A2A receptor antagonist.
2378. The device of item 2376 wherein the agent is an AKT inhibitor.
2379. The device of item 2376 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
2380. The device of item 2376 wherein the agent is an alpha 4 integrin antagonist.
2381. The device of item 2376 wherein the agent is an alpha 7 nicotinic receptor agonist.
2382. The device of item 2376 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- , 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2383. The device of item 2376 wherein the agent is an apoptosis antagonist.
2384. The device of item 2376 wherein the agent is an apoptosis activator.
2385. The device of item 2376 wherein the agent is a beta 1 integrin antagonist.
2386. The device of item 2376 wherein the agent is a beta tubulin inhibitor. 2387. The device of item 2376 wherein the agent is a blocker of enzyme production in Hepatitis C.
2388. The device of item 2376 wherein the agent is a Bruton's tyrosine kinase inhibitor.
2389. The device of item 2376 wherein the agent is a calcineurin inhibitor.
2390. The device of item 2376 wherein the agent is a caspase 3 inhibitor.
2391. The device of item 2376 wherein the agent is a CC chemokine receptor antagonist.
2392. The device of item 2376 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2393. The device of item 2376 wherein the agent is a cathepsin B inhibitor.
2394. The device of item 2376 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
2395. The device of item 2376 wherein the agent is a cathepsin L inhibitor.
2396. The device of item 2376 wherein the agent is a CD40 antagonist.
2397. The device of item 2376 wherein the agent is a chemokine receptor agonist. 2398. The device of item 2376 wherein the agent is a chymase inhibitor.
2399. The device of item 2376 wherein the agent is a collagenase antagonist.
2400. The device of item 2376 wherein the agent is a CXCR antagonist.
2401. The device of item 2376 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2402. The device of item 2376 wherein the agent is a cyclooxygenase 1 inhibitor.
2403. The device of item 2376 wherein the agent is a DHFR inhibitor.
2404. The device of item 2376 wherein the agent is a dual integrin inhibitor.
2405. The device of item 2376 wherein the agent is an elastase inhibitor.
2406. The device of item 2376 wherein the agent is an elongation factor-1 alpha inhibitor. 2407. The device of item 2376 wherein the agent is an endothelial growth factor antagonist.
2408. The device of item 2376 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2409. The device of item 2376 wherein the agent is an endotoxin antagonist.
2410. The device of item 2376 wherein the agent is an epothilone and tubulin binder.
2411. The device of item 2376 wherein the agent is an estrogen receptor antagonist.
2412. The device of item 2376 wherein the agent is an FGF inhibitor.
2413. The device of item 2376 wherein the agent is a farnexyl transferase inhibitor.
2414. The device of item 2376 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2415. The device of item 2376 wherein the agent is an FLT-3 kinase inhibitor.
2416. The device of item 2376 wherein the agent is an FGF receptor kinase inhibitor.
2417. The device of item 2376 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
2418. The device of item 2376 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
2419. The device of item 2376 wherein the agent is a histone deacetylase inhibitor.
2420. The device of item 2376 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 2421. The device of item 2376 wherein the agent is an ICAM inhibitor.
2422. The device of item 2376 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2423. The device of item 2376 wherein the agent is an IL-2 inhibitor.
2424. The device of item 2376 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
2425. The device of item 2376 wherein the agent is an IMPDH (inosine monophosphate). 2426. The device of item 2376 wherein the agent is an integrin antagonist.
2427. The device of item 2376 wherein the agent is an interleukin antagonist.
2428. The device of item 2376 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
2429. The device of item 2376 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
2430. The device of item 2376 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
2431. The device of item 2376 wherein the agent a JAK3 enzyme inhibitor.
2432. The device of item 2376 wherein the agent is a JNK inhibitor.
2433. The device of item 2376 wherein the agent is a kinase inhibitor.
2434. The device of item 2376 wherein the agent is kinesin antagonist.
2435. The device of item 2376 wherein the agent is a kinesin antagonist.
2436. The device of item 2376 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2437. The device of item 2376 wherein the agent is an MAP kinase inhibitor.
2438. The device of item 2376 wherein the agent is a matrix metalloproteinase inhibitor.
2439. The device of item 2376 wherein the agent is an MCP-
CCR2 inhibitor.
2440. The device of item 2376 wherein the agent is an mTOR inhibitor.
2441. The device of item 2376 wherein the agent is an mTOR kinase inhibitor.
2442. The device of item 2376 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
2443. The device of item 2376 wherein the agent is an MIF inhibitor.
2444. The device of item 2376 wherein the agent is an MMP inhibitor.
2445. The device of item 2376 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2446. The device of item 2376 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 2447. The device of item 2376 wherein the agent is a nitric oxide agonist.
2448. The device of item 2376 wherein the agent is an ornithine decarboxylase inhibitor.
2449. The device of item 2376 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
2450. The device of item 2376 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
2451. The device of item 2376 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2452. The device of item 2376 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKiine), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKiine), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2453. The device of item 2376 wherein the agent is a phosphatase inhibitor.
2454. The device of item 2376 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
2455. The device of item 2376 wherein the agent is a PKC inhibitor.
2456. The device of item 2376 wherein the agent is a platelet activating factor antagonist.
2457. The device of item 2376 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2458. The device of item 2376 wherein the agent is a prolyl hydroxylase inhibitor.
2459. The device of item 2376 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2460. The device of item 2376 wherein the agent is a protein kinase B inhibitor.
2461. The device of item 2376 wherein the agent is a protein kinase C stimulant.
2462. The device of item 2376 wherein the agent is a purine nucleoside analogue.
2463. The device of item 2376 wherein the agent is a purinoreceptor P2X antagonist.
2464. The device of item 2376 wherein the agent is a Raf kinase inhibitor. 2465. The device of item 2376 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2466. The device of item 2376 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2467. The device of item 2376 wherein the agent is an SDF-1 antagonist.
2468. The device of item 2376 wherein the agent is a sheddase inhibitor.
2469. The device of item 2376 wherein the agent is an SRC inhibitor.
2470. The device of item 2376 wherein the agent is a stromelysin inhibitor.
2471. The device of item 2376 wherein the agent is an Syk kinase inhibitor.
2472. The device of item 2376 wherein the agent is a telomerase inhibitor.
2473. The device of item 2376 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme
(Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
2474. The device of item 2376 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2475. The device of item 2376 wherein the agent is a Toll receptor inhibitor.
2476. The device of item 2376 wherein the agent is a tubulin antagonist.
2477. The device of item 2376 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2478. The device of item 2376 wherein the agent is a VEGF inhibitor.
2479. The device of item 2376 wherein the agent is a vitamin D receptor agonist.
2480. The device of item 2376 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
2481. The device of item 2376 wherein the agent is AP-23573 (an mTOR inhibitor). 2482. The device of item 2376 wherein the agent is synthadotin (a tubulin antagonist).
2483. The device of item 2376 wherein the agent is S-0885 (a collagenase inhibitor).
2484. The device of item 2376 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2485. The device of item 2376 wherein the agent is ixabepilone (an epithilone).
2486. The device of item 2376 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2487. The device of item 2376 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2488. The device of item 2376 wherein the agent is ABT-518 (an angiogenesis inhibitor).
2489. The device of item 2376 wherein the agent is combretastatin (an angiogenesis inhibitor).
2490. The device of item 2376 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2491. The device of item 2376 wherein the agent is SB-715992 (a kinesin antagonist).
2492. The device of item 2376 wherein the agent is temsirolimus (an mTOR inhibitor).
2493. The device of item 2376 wherein the agent is adalimumab (a TNFα antagonist). 2494. The device of item 2376, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
2495. The device of item 2376, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2496. The device of item 2376, further comprising a coating, wherein the coating is disposed on a surface of the device.
2497. The device of item 2376, further comprising a coating, wherein the coating directly contacts the device.
2498. The device of item 2376, further comprising a coating, wherein the coating indirectly contacts the device.
2499. The device of item 2376, further comprising a coating, wherein the coating partially covers the device.
2500. The device of item 2376, further comprising a coating, wherein the coating completely covers the device.
2501. The device of item 2376, further comprising a coating, wherein the coating is a uniform coating.
2502. The device of item 2376, further comprising a coating, wherein the coating is a non-uniform coating.
2503. The device of item 2376, further comprising a coating, wherein the coating is a discontinuous coating.
2504. The device of item 2376, further comprising a coating, wherein the coating is a patterned coating.
2505. The device of item 2376, further comprising a coating, wherein the coating has a thickness of 100 μm or less. 2506. The device of item 2376, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
2507. The device of item 2376, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2508. The device of item 2376, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2509. The device of item 2376, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
2510. The device of item 2376, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2511. The device of item 2376, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2512. The device of item 2376, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2513. The device of item 2376, further comprising a coating, wherein the coating further comprises a polymer.
2514. The device of item 2376, further comprising a first coating having a first composition and the second coating having a second composition.
2515. The device of item 2376, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2516. The device of item 2376, further comprising a polymer.
2517. The device of item 2376, further comprising a polymeric carrier.
2518. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2519. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2520. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2521. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2522. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2523. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2524. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2525. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2526. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains. 2527. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2528. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2529. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2530. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2531. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2532. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2533. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2534. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2535. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2536. The device of item 2376, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2537. The device of item 2376, further comprising a lubricious coating.
2538. The device of item 2376 wherein the anti-scarring agent is located within pores or holes of the device. 2539. The device of item 2376 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2540. The device of item 2376, further comprising a second pharmaceutically active agent.
2541. The device of item 2376, further comprising an antiinflammatory agent.
2542. The device of item 2376, further comprising an agent that inhibits infection.
2543. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2544. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2545. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2546. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2547. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2548. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2549. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2550. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin. 2551. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2552. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2553. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2554. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2555. The device of item 2376, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2556. The device of item 2376, further comprising an antithrombotic agent.
2557. The device of item 2376, further comprising a visualization agent.
2558. The device of item 2376, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2559. The device of item 2376, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2560. The device of item 2376, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material. 2561. The device of item 2376, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2562. The device of item 2376, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2563. The device of item 2376, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2564. The device of item 2376, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2565. The device of item 2376, further comprising an echogenic material.
2566. The device of item 2376, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2567. The device of item 2376 wherein the device is sterile.
2568. The device of item 2376 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2569. The device of item 2376 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
2570. The device of item 2376 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. 2571. The device of item 2376 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2572. The device of item 2376 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2573. The device of item 2376 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2574. The device of item 2376 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2575. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2576. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2577. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2578. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2579. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 2580. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2581. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2582. The device of item 2376 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2583. The device of item 2376 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
2584. The device of item 2376 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
2585. The device of item 2376 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2586. The device of item 2376 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2587. The device of item 2376 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2588. The device of item 2376 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm of device surface to which the anti-scarring agent is applied.
2589. The device of item 2376 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2590. The device of item 2376 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2591. The device of item 2376 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2592. The device of item 2376 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2593. The device of item 2376 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2594. The device of item 2376 wherein the agent or the composition is affixed to the sensor.
2595. The device of item 2376 wherein the agent or the composition is covalently attached to the sensor.
2596. The device of item 2376 wherein the agent or the composition is non-covalently attached to the sensor.
2597. The device of item 2376 further comprising a coating that absorbs the agent or the composition.
2598. The device of item 2376 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
2599. The device of item 2376 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition. 2600. The device of item 2376 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
2601. The device of item 2376 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
2602. The device of item 2376 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
2603. The device of item 2376 further comprising a pump that is linked to the sensor.
2604. The device of any one of items 2376-2603 wherein the device is adapted for delivering 2'-deoxy 5-fluorouridine.
2605. The device of any one of items 2376-2603 wherein the host has a solid tumor, and the device is adapted for infusing a chemotherapeutic agent to the solid tumor.
2606. The device of any one of items 2376-2603 wherein the host has a tumor, and the device is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
2607. The device of any one of items 2376-2603 wherein the host has a hepatic tumor, and the device is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host.
2608. A device, comprising a drug delivery pump for treating heart disease and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2609. The device of item 2608 wherein the agent is an adensosine A2A receptor antagonist. 2610. The device of item 2608 wherein the agent is an AKT inhibitor.
2611. The device of item 2608 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
2612. The device of item 2608 wherein the agent is an alpha 4 integrin antagonist.
2613. The device of item 2608 wherein the agent is an alpha 7 nicotinic receptor agonist.
2614. The device of item 2608 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attention), WX-293 (Wilex), M-2025 (Metris Therapeutics), Aiphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2615. The device of item 2608 wherein the agent is an apoptosis antagonist.
2616. The device of item 2608 wherein the agent is an apoptosis activator.
2617. The device of item 2608 wherein the agent is a beta 1 integrin antagonist.
2618. The device of item 2608 wherein the agent is a beta tubulin inhibitor.
2619. The device of item 2608 wherein the agent is a blocker of enzyme production in Hepatitis C.
2620. The device of item 2608 wherein the agent is a Bruton's tyrosine kinase inhibitor.
2621. The device of item 2608 wherein the agent is a calcineurin inhibitor.
2622. The device of item 2608 wherein the agent is a caspase 3 inhibitor. 2623. The device of item 2608 wherein the agent is a CC chemokine receptor antagonist.
2624. The device of item 2608 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2625. The device of item 2608 wherein the agent is a cathepsin B inhibitor.
2626. The device of item 2608 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
2627. The device of item 2608 wherein the agent is a cathepsin L inhibitor.
2628. The device of item 2608 wherein the agent is a CD40 antagonist.
2629. The device of item 2608 wherein the agent is a chemokine receptor agonist.
2630. The device of item 2608 wherein the agent is a chymase inhibitor.
2631. The device of item 2608 wherein the agent is a collagenase antagonist.
2632. The device of item 2608 wherein the agent is a CXCR antagonist.
2633. The device of item 2608 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2634. The device of item 2608 wherein the agent is a cyclooxygenase 1 inhibitor.
2635. The device of item 2608 wherein the agent is a DHFR inhibitor.
2636. The device of item 2608 wherein the agent is a dual integrin inhibitor.
2637. The device of item 2608 wherein the agent is an elastase inhibitor.
2638. The device of item 2608 wherein the agent is an elongation factor-1 alpha inhibitor.
2639. The device of item 2608 wherein the agent is an endothelial growth factor antagonist.
2640. The device of item 2608 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2641. The device of item 2608 wherein the agent is an endotoxin antagonist.
2642. The device of item 2608 wherein the agent is an epothilone and tubulin binder.
2643. The device of item 2608 wherein the agent is an estrogen receptor antagonist.
2644. The device of item 2608 wherein the agent is an FGF inhibitor.
2645. The device of item 2608 wherein the agent is a famexyl transferase inhibitor.
2646. The device of item 2608 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2647. The device of item 2608 wherein the agent is an FLT-3 kinase inhibitor.
2648. The device of item 2608 wherein the agent is an FGF receptor kinase inhibitor.
2649. The device of item 2608 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-
13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
2650. The device of item 2608 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
2651. The device of item 2608 wherein the agent is a hϊstone deacetylase inhibitor.
2652. The device of item 2608 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
2653. The device of item 2608 wherein the agent is an ICAM inhibitor.
2654. The device of item 2608 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2655. The device of item 2608 wherein the agent is an IL-2 inhibitor.
2656. The device of item 2608 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
2657. The device of item 2608 wherein the agent is an IMPDH (inosine monophosphate).
2658. The device of item 2608 wherein the agent is an integrin antagonist.
2659. The device of item 2608 wherein the agent is an interleukin antagonist.
2660. The device of item 2608 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
2661. The device of item 2608 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
2662. The device of item 2608 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 2663. The device of item 2608 wherein the agent a JAK3 enzyme inhibitor.
2664. The device of item 2608 wherein the agent is a JNK inhibitor.
2665. The device of item 2608 wherein the agent is a kinase inhibitor.
2666. The device of item 2608 wherein the agent is kinesin antagonist.
2667. The device of item 2608 wherein the agent is a kinesin antagonist.
2668. The device of item 2608 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2669. The device of item 2608 wherein the agent is an MAP kinase inhibitor. 2670. The device of item 2608 wherein the agent is a matrix metalloproteinase inhibitor.
2671. The device of item 2608 wherein the agent is an MCP- CCR2 inhibitor.
2672. The device of item 2608 wherein the agent is an mTOR inhibitor.
2673. The device of item 2608 wherein the agent is an mTOR kinase inhibitor.
2674. The device of item 2608 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
2675. The device of item 2608 wherein the agent is an MIF inhibitor.
2676. The device of item 2608 wherein the agent is an MMP inhibitor.
2677. The device of item 2608 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2678. The device of item 2608 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
2679. The device of item 2608 wherein the agent is a nitric oxide agonist.
2680. The device of item 2608 wherein the agent is an ornithine decarboxylase inhibitor.
2681. The device of item 2608 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof. 2682. The device of item 2608 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
2683. The device of item 2608 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2684. The device of item 2608 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2685. The device of item 2608 wherein the agent is a phosphatase inhibitor.
2686. The device of item 2608 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
2687. The device of item 2608 wherein the agent is a PKC inhibitor.
2688. The device of item 2608 wherein the agent is a platelet activating factor antagonist. 2689. The device of item 2608 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2690. The device of item 2608 wherein the agent is a prolyl hydroxylase inhibitor.
2691. The device of item 2608 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2692. The device of item 2608 wherein the agent is a protein kinase B inhibitor.
2693. The device of item 2608 wherein the agent is a protein kinase C stimulant.
2694. The device of item 2608 wherein the agent is a purine nucleoside analogue.
2695. The device of item 2608 wherein the agent is a purinoreceptor P2X antagonist.
2696. The device of item 2608 wherein the agent is a Raf kinase inhibitor.
2697. The device of item 2608 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2698. The device of item 2608 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2699. The device of item 2608 wherein the agent is an SDF-1 antagonist.
2700. The device of item 2608 wherein the agent is a sheddase inhibitor. 2701. The device of item 2608 wherein the agent is an SRC inhibitor.
2702. The device of item 2608 wherein the agent is a stromelysin inhibitor.
2703. The device of item 2608 wherein the agent is an Syk kinase inhibitor.
2704. The device of item 2608 wherein the agent is a telomerase inhibitor.
2705. The device of item 2608 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-
8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
2706. The device of item 2608 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2707. The device of item 2608 wherein the agent is a Toll receptor inhibitor.
2708. The device of item 2608 wherein the agent is a tubulin antagonist. 2709. The device of item 2608 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsϊan Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2710. The device of item 2608 wherein the agent is a VEGF inhibitor.
2711. The device of item 2608 wherein the agent is a vitamin D receptor agonist.
2712. The device of item 2608 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
2713. The device of item 2608 wherein the agent is AP-23573 (an mTOR inhibitor).
2714. The device of item 2608 wherein the agent is synthadotin (a tubulin antagonist).
2715. The device of item 2608 wherein the agent is S-0885 (a collagenase inhibitor).
2716. The device of item 2608 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2717. The device of item 2608 wherein the agent is ixabepilone
(an epithilone). 2718. The device of item 2608 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2719. The device of item 2608 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2720. The device of item 2608 wherein the agent is ABT-518
(an angiogenesis inhibitor).
2721. The device of item 2608 wherein the agent is combretastatin (an angiogenesis inhibitor).
2722. The device of item 2608 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2723. The device of item 2608 wherein the agent is SB-715992 (a kinesin antagonist).
2724. The device of item 2608 wherein the agent is temsirolimus (an mTOR inhibitor).
2725. The device of item 2608 wherein the agent is adalimumab (a TNFα antagonist).
2726. The device of item 2608, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
2727. The device of item 2608, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2728. The device of item 2608, further comprising a coating, wherein the coating is disposed on a surface of the device.
2729. The device of item 2608, further comprising a coating, wherein the coating directly contacts the device. 2730. The device of item 2608, further comprising a coating, wherein the coating indirectly contacts the device.
2731. The device of item 2608, further comprising a coating, wherein the coating partially covers the device.
2732. The device of item 2608, further comprising a coating, wherein the coating completely covers the device.
2733. The device of item 2608, further comprising a coating, wherein the coating is a uniform coating.
2734. The device of item 2608, further comprising a coating, wherein the coating is a non-uniform coating.
2735. The device of item 2608, further comprising a coating, wherein the coating is a discontinuous coating.
2736. The device of item 2608, further comprising a coating, wherein the coating is a patterned coating.
2737. The device of item 2608, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
2738. The device of item 2608, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
2739. The device of item 2608, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2740. The device of item 2608, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year. 2741. The device of item 2608, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2742. The device of item 2608, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2743. The device of item 2608, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2744. The device of item 2608, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2745. The device of item 2608, further comprising a coating, wherein the coating further comprises a polymer.
2746. The device of item 2608, further comprising a first coating having a first composition and the second coating having a second composition.
2747. The device of item 2608, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2748. The device of item 2608, further comprising a polymer.
2749. The device of item 2608, further comprising a polymeric carrier.
2750. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer. 2751. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2752. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2753. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2754. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2755. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2756. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2757. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2758. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2759. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2760. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2761. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel. 2762. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2763. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2764. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2765. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2766. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2767. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2768. The device of item 2608, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2769. The device of item 2608, further comprising a lubricious coating.
2770. The device of item 2608 wherein the anti-scarring agent is located within pores or holes of the device.
2771. The device of item 2608 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2772. The device of item 2608, further comprising a second pharmaceutically active agent.
2773. The device of item 2608, further comprising an anti- inflammatory agent. 2774. The device of item 2608, further comprising an agent that inhibits infection.
2775. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2776. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2777. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2778. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2779. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2780. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2781. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2782. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2783. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2784. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2785. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 2786. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2787. The device of item 2608, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2788. The device of item 2608, further comprising an antithrombotic agent.
2789. The device of item 2608, further comprising a visualization agent.
2790. The device of item 2608, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2791. The device of item 2608, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2792. The device of item 2608, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2793. The device of item 2608, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2794. The device of item 2608, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 2795. The device of item 2608, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2796. The device of item 2608, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2797. The device of item 2608, further comprising an echogenic material.
2798. The device of item 2608, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2799. The device of item 2608 wherein the device is sterile.
2800. The device of item 2608 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2801. The device of item 2608 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
2802. The device of item 2608 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2803. The device of item 2608 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2804. The device of item 2608 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue. 2805. The device of item 2608 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2806. The device of item 2608 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2807. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2808. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2809. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2810. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2811. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2812. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2813. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 2814. The device of item 2608 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2815. The device of item 2608 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
2816. The device of item 2608 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
2817. The device of item 2608 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2818. The device of item 2608 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2819. The device of item 2608 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2820. The device of item 2608 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2821. The device of item 2608 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2822. The device of item 2608 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2823. The device of item 2608 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2824. The device of item 2608 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2825. The device of item 2608 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2826. The device of item 2608 wherein the agent or the composition is affixed to the sensor.
2827. The device of item 2608 wherein the agent or the composition is covalently attached to the sensor.
2828. The device of item 2608 wherein the agent or the composition is non-covalently attached to the sensor.
2829. The device of item 2608 further comprising a coating that absorbs the agent or the composition.
2830. The device of item 2608 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
2831. The device of item 2608 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
2832. The device of item 2608 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
2833. The device of item 2608 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
2834. The device of item 2608 wherein the sensor is completely covered with a mesh that contains the agent or the composition. 2835. The device of item 2608 further comprising a pump that is linked to the sensor.
2836. The device of any one of items 2608-2835 wherein the device is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site.
2837. A device, comprising a drug delivery implant (i.e., a pump) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2838. The device of item 2837 wherein the agent is an adensosine A2A receptor antagonist.
2839. The device of item 2837 wherein the agent is an AKT inhibitor.
2840. The device of item 2837 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects
No. 5754 (Merck KgaA).
2841. The device of item 2837 wherein the agent is an alpha 4 integrin antagonist.
2842. The device of item 2837 wherein the agent is an alpha 7 nicotinic receptor agonist.
2843. The device of item 2837 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTl-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant
Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732
(Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-
23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2844. The device of item 2837 wherein the agent is an apoptosis antagonist.
2845. The device of item 2837 wherein the agent is an apoptosis activator.
ι 2846. The device of item 2837 wherein the agent is a beta 1 integrin antagonist. 2847. The device of item 2837 wherein the agent is a beta tubulin inhibitor.
2848. The device of item 2837 wherein the agent is a blocker of enzyme production in Hepatitis C.
2849. The device of item 2837 wherein the agent is a Bruton's tyrosine kinase inhibitor.
2850. The device of item 2837 wherein the agent is a calcineurin inhibitor.
2851. The device of item 2837 wherein the agent is a caspase 3 inhibitor.
2852. The device of item 2837 wherein the agent is a CC chemokine receptor antagonist.
2853. The device of item 2837 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2854. The device of item 2837 wherein the agent is a cathepsin B inhibitor.
2855. The device of item 2837 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
2856. The device of item 2837 wherein the agent is a cathepsin L inhibitor.
2857. The device of item 2837 wherein the agent is a CD40 antagonist. 2858. The device of item 2837 wherein the agent is a chemokine receptor agonist.
2859. The device of item 2837 wherein the agent is a chymase inhibitor.
2860. The device of item 2837 wherein the agent is a collagenase antagonist.
2861. The device of item 2837 wherein the agent is a CXCR antagonist.
2862. The device of item 2837 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2863. The device of item 2837 wherein the agent is a cyclooxygenase 1 inhibitor.
2864. The device of item 2837 wherein the agent is a DHFR inhibitor.
2865. The device of item 2837 wherein the agent is a dual integrin inhibitor.
2866. The device of item 2837 wherein the agent is an elastase inhibitor. 2867. The device of item 2837 wherein the agent is an elongation factor-1 alpha inhibitor.
2868. The device of item 2837 wherein the agent is an endothelial growth factor antagonist.
2869. The device of item 2837 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2870. The device of item 2837 wherein the agent is an endotoxin antagonist.
2871. The device of item 2837 wherein the agent is an epothilone and tubulin binder.
2872. The device of item 2837 wherein the agent is an estrogen receptor antagonist.
2873. The device of item 2837 wherein the agent is an FGF inhibitor.
2874. The device of item 2837 wherein the agent is a farnexyl transferase inhibitor.
2875. The device of item 2837 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2876. The device of item 2837 wherein the agent is an FLT-3 kinase inhibitor.
2877. The device of item 2837 wherein the agent is an FGF receptor kinase inhibitor.
2878. The device of item 2837 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-
13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
2879. The device of item 2837 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
2880. The device of item 2837 wherein the agent is a histone deacetylase inhibitor.
2881. The device of item 2837 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 2882. The device of item 2837 wherein the agent is an ICAM inhibitor.
2883. The device of item 2837 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2884. The device of item 2837 wherein the agent is an IL-2 inhibitor.
2885. The device of item 2837 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
2886. The device of item 2837 wherein the agent is an IMPDH (inosine monophosphate). 2887. The device of item 2837 wherein the agent is an integrin antagonist.
2888. The device of item 2837 wherein the agent is an interleukin antagonist.
2889. The device of item 2837 wherein the agent is an inhibitor of type Hf receptor tyrosine kinase.
2890. The device of item 2837 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
2891. The device of item 2837 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
2892. The device of item 2837 wherein the agent a JAK3 enzyme inhibitor.
2893. The device of item 2837 wherein the agent is a JNK inhibitor.
2894. The device of item 2837 wherein the agent is a kinase inhibitor.
2895. The device of item 2837 wherein the agent is kinesin antagonist.
2896. The device of item 2837 wherein the agent is a kinesin antagonist.
2897. The device of item 2837 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119D340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2898. The device of item 2837 wherein the agent is an MAP kinase inhibitor.
2899. The device of item 2837 wherein the agent is a matrix metalloproteinase inhibitor.
2900. The device of item 2837 wherein the agent is an MCP-
CCR2 inhibitor.
2901. The device of item 2837 wherein the agent is an mTOR inhibitor.
2902. The device of item 2837 wherein the agent is an mTOR kinase inhibitor.
2903. The device of item 2837 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
2904. The device of item 2837 wherein the agent is an MIF inhibitor.
2905. The device of item 2837 wherein the agent is an MMP inhibitor.
2906. The device of item 2837 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2907. The device of item 2837 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 2908. The device of item 2837 wherein the agent is a nitric oxide agonist.
2909. The device of item 2837 wherein the agent is an ornithine decarboxylase inhibitor.
2910. The device of item 2837 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
2911. The device of item 2837 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
2912. The device of item 2837 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vataϊanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2913. The device of item 2837 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2914. The device of item 2837 wherein the agent is a phosphatase inhibitor.
2915. The device of item 2837 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
2916. The device of item 2837 wherein the agent is a PKC inhibitor.
2917. The device of item 2837 wherein the agent is a platelet activating factor antagonist.
2918. The device of item 2837 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2919. The device of item 2837 wherein the agent is a prolyl hydroxylase inhibitor.
2920. The device of item 2837 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2921. The device of item 2837 wherein the agent is a protein kinase B inhibitor.
2922. The device of item 2837 wherein the agent is a protein kinase C stimulant.
2923. The device of item 2837 wherein the agent is a purine nucleoside analogue.
2924. The device of item 2837 wherein the agent is a purinoreceptor P2X antagonist.
2925. The device of item 2837 wherein the agent is a Raf kinase inhibitor. 2926. The device of item 2837 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2927. The device of item 2837 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2928. The device of item 2837 wherein the agent is an SDF-1 antagonist.
2929. The device of item 2837 wherein the agent is a sheddase inhibitor.
2930. The device of item 2837 wherein the agent is an SRC inhibitor.
2931. The device of item 2837 wherein the agent is a stromelysin inhibitor.
2932. The device of item 2837 wherein the agent is an Syk kinase inhibitor.
2933. The device of item 2837 wherein the agent is a telomerase inhibitor.
2934. The device of item 2837 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13- 8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme
(Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
2935. The device of item 2837 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegoi (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2936. The device of item 2837 wherein the agent is a Toll receptor inhibitor.
2937. The device of item 2837 wherein the agent is a tubulin antagonist.
2938. The device of item 2837 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, 1)3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2939. The device of item 2837 wherein the agent is a VEGF inhibitor.
2940. The device of item 2837 wherein the agent is a vitamin D receptor agonist.
2941. The device of item 2837 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
2942. The device of item 2837 wherein the agent is AP-23573 (an mTOR inhibitor). 2943. The device of item 2837 wherein the agent is synthadotin (a tubulin antagonist).
2944. The device of item 2837 wherein the agent is S-0885 (a collagenase inhibitor).
2945. The device of item 2837 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2946. The device of item 2837 wherein the agent is ixabepilone (an epithilone).
2947. The device of item 2837 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2948. The device of item 2837 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2949. The device of item 2837 wherein the agent is ABT-518 (an angiogenesis inhibitor).
2950. The device of item 2837 wherein the agent is combretastatin (an angiogenesis inhibitor).
2951. The device of item 2837 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2952. The device of item 2837 wherein the agent is SB-715992 (a kinesin antagonist).
2953. The device of item 2837 wherein the agent is temsirolimus (an mTOR inhibitor).
2954. The device of item 2837 wherein the agent is adalimumab (a TNFα antagonist). 2955. The device of item 2837, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
2956. The device of item 2837, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2957. The device of item 2837, further comprising a coating, wherein the coating is disposed on a surface of the device.
2958. The device of item 2837, further comprising a coating, wherein the coating directly contacts the device.
2959. The device of item 2837, further comprising a coating, wherein the coating indirectly contacts the device.
2960. The device of item 2837, further comprising a coating, wherein the coating partially covers the device.
2961. The device of item 2837, further comprising a coating, wherein the coating completely covers the device.
2962. The device of item 2837, further comprising a coating, wherein the coating is a uniform coating.
2963. The device of item 2837, further comprising a coating, wherein the coating is a non-uniform coating.
2964. The device of item 2837, further comprising a coating, wherein the coating is a discontinuous coating.
2965. The device of item 2837, further comprising a coating, wherein the coating is a patterned coating.
2966. The device of item 2837, further comprising a coating, wherein the coating has a thickness of 100 μm or less. 2967. The device of item 2837, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
2968. The device of item 2837, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2969. The device of item 2837, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2970. The device of item 2837, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
2971. The device of item 2837, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2972. The device of item 2837, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2973. The device of item 2837, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2974. The device of item 2837, further comprising a coating, wherein the coating further comprises a polymer.
2975. The device of item 2837, further comprising a first coating having a first composition and the second coating having a second composition.
2976. The device of item 2837, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2977. The device of item 2837, further comprising a polymer.
2978. The device of item 2837, further comprising a polymeric carrier.
2979. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2980. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2981. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2982. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2983. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2984. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2985. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2986. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2987. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains. 2988. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2989. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2990. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2991. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2992. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2993. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2994. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2995. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2996. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2997. The device of item 2837, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2998. The device of item 2837, further comprising a lubricious coating.
2999. The device of item 2837 wherein the anti-scarring agent is located within pores or holes of the device. 3000. The device of item 2837 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3001. The device of item 2837, further comprising a second pharmaceutically active agent.
3002. The device of item 2837, further comprising an antiinflammatory agent.
3003. The device of item 2837, further comprising an agent that inhibits infection.
3004. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
3005. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3006. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3007. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3008. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3009. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3010. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3011. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin. 3012. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3013. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3014. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
3015. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3016. The device of item 2837, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3017. The device of item 2837, further comprising an antithrombotic agent.
3018. The device of item 2837, further comprising a visualization agent.
3019. The device of item 2837, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3020. The device of item 2837, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3021. The device of item 2837, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material. 3022. The device of item 2837, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
3023. The device of item 2837, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3024. The device of item 2837, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3025. The device of item 2837, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3026. The device of item 2837, further comprising an echogenic material.
3027. The device of item 2837, further comprising an echogenic
material, wherein the echogenic material is in the form of a coating.
3028. The device of item 2837 wherein the device is sterile.
3029. The device of item 2837 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
3030. The device of item 2837 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
3031. The device of item 2837 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. 3032. The device of item 2837 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
3033. The device of item 2837 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3034. The device of item 2837 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3035. The device of item 2837 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3036. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3037. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3038. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3039. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3040. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 3041. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3042. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3043. The device of item 2837 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3044. The device of item 2837 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
3045. The device of item 2837 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
3046. The device of item 2837 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3047. The device of item 2837 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3048. The device of item 2837 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3049. The device of item 2837 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3050. The device of item 2837 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 3051. The device of item 2837 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3052. The device of item 2837 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3053. The device of item 2837 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3054. The device of item 2837 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3055. The device of item 2837 wherein the agent or the composition is affixed to the sensor.
3056. The device of item 2837 wherein the agent or the composition is covalently attached to the sensor.
3057. The device of item 2837 wherein the agent or the composition is non-covalently attached to the sensor.
3058. The device of item 2837 further comprising a coating that absorbs the agent or the composition.
3059. The device of item 2837 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
3060. The device of item 2837 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition. 3061. The device of item 2837 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
3062. The device of item 2837 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
3063. The device of item 2837 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
3064. The device of item 2837 further comprising a pump that is linked to the sensor.
3065. A method for inhibiting scarring comprising placing a sensor and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
3066. The method of item 3065 wherein the agent is an adensosine A2A receptor antagonist.
3067. The method of item 3065 wherein the agent is an AKT inhibitor.
3068. The method of item 3065 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3069. The method of item 3065 wherein the agent is an alpha 4 integrin antagonist.
3070. The method of item 3065 wherein the agent is an alpha 7 nicotinic receptor agonist.
3071. The method of item 3065 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3072. The method of item 3065 wherein the agent is an apoptosis antagonist.
3073. The method of item 3065 wherein the agent is an apoptosis activator. 3074. The method of item 3065 wherein the agent is a beta 1 integrin antagonist.
3075. The method of item 3065 wherein the agent is a beta tubulin inhibitor.
3076. The method of item 3065 wherein the agent is a blocker of enzyme production in Hepatitis C.
3077. The method of item 3065 wherein the agent is a Bruton's tyrosine kinase inhibitor.
3078. The method of item 3065 wherein the agent is a calcineurin inhibitor.
3079. The method of item 3065 wherein the agent is a caspase 3 inhibitor.
3080. The method of item 3065 wherein the agent is a CC chemokine receptor antagonist.
3081. The method of item 3065 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3082. The method of item 3065 wherein the agent is a cathepsin B inhibitor.
3083. The method of item 3065 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
3084. The method of item 3065 wherein the agent is a cathepsin L inhibitor. 3085. The method of item 3065 wherein the agent is a CD40 antagonist.
3086. The method of item 3065 wherein the agent is a chemokine receptor agonist.
3087. The method of item 3065 wherein the agent is a chymase inhibitor.
3088. The method of item 3065 wherein the agent is a collagenase antagonist.
3089. The method of item 3065 wherein the agent is a CXCR antagonist.
3090. The method of item 3065 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3091. The method of item 3065 wherein the agent is a cyclooxygenase 1 inhibitor.
3092. The method of item 3065 wherein the agent is a DHFR inhibitor.
3093. The method of item 3065 wherein the agent is a dual integrin inhibitor. 3094. The method of item 3065 wherein the agent is an elastase inhibitor.
3095. The method of item 3065 wherein the agent is an elongation factor-1 alpha inhibitor.
3096. The method of item 3065 wherein the agent is an endothelial growth factor antagonist.
3097. The method of item 3065 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB)1 KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3098. The method of item 3065 wherein the agent is an endotoxin antagonist.
3099. The method of item 3065 wherein the agent is an epothilone and tubulin binder.
3100. The method of item 3065 wherein the agent is an estrogen receptor antagonist.
3101. The method of item 3065 wherein the agent is an FGF inhibitor.
3102. The method of item 3065 wherein the agent is a farnexyl transferase inhibitor. 3103. The method of item 3065 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIIl (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3104. The method of item 3065 wherein the agent is an FLT-3 kinase inhibitor.
3105. The method of item 3065 wherein the agent is an FGF receptor kinase inhibitor.
3106. The method of item 3065 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3107. The method of item 3065 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
3108. The method of item 3065 wherein the agent is a histone deacetylase inhibitor.
3109. The method of item 3065 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
3110. The method of item 3065 wherein the agent is an ICAM inhibitor.
3111. The method of item 3065 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
3112. The method of item 3065 wherein the agent is an IL-2 inhibitor.
3113. The method of item 3065 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-O01 (Protalex), and an analogue or derivative thereof. 3114. The method of item 3065 wherein the agent is an IMPDH (inosine monophosphate).
3115. The method of item 3065 wherein the agent is an integrin antagonist.
3116. The method of item 3065 wherein the agent is an interleukin antagonist.
3117. The method of item 3065 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3118. The method of item 3065 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3119. The method of item 3065 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
3120. The method of item 3065 wherein the agent a JAK3 enzyme inhibitor.
3121. The method of item 3065 wherein the agent is a JNK inhibitor. . >
3122. The method of item 3065 wherein the agent is a kinase inhibitor.
3123. The method of item 3065 wherein the agent is kinesin antagonist.
3124. The method of item 3065 wherein the agent is a kinesin antagonist.
3125. The method of item 3065 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3126. The method of item 3065 wherein the agent is an MAP kinase inhibitor.
3127. The method of item 3065 wherein the agent is a matrix metalloproteinase inhibitor.
3128. The method of item 3065 wherein the agent is an MCP- CCR2 inhibitor.
3129. The method of item 3065 wherein the agent is an mTOR inhibitor.
3130. The method of item 3065 wherein the agent is an mTOR kinase inhibitor.
3131. The method of item 3065 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
3132. The method of item 3065 wherein the agent is an MIF inhibitor.
3133. The method of item 3065 wherein the agent is an MMP inhibitor.
3134. The method of item 3065 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
3135. The method of item 3065 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3136. The method of item 3065 wherein the agent is a nitric oxide agonist.
3137. The method of item 3065 wherein the agent is an ornithine decarboxylase inhibitor.
3138. The method of item 3065 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3139. The method of item 3065 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3140. The method of item 3065 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3141. The method of item 3065 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3142. The method of item 3065 wherein the agent is a phosphatase inhibitor.
3143. The method of item 3065 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase ill inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3144. The method of item 3065 wherein the agent is a PKC inhibitor.
3145. The method of item 3065 wherein the agent is a platelet activating factor antagonist.
3146. The method of item 3065 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3147. The method of item 3065 wherein the agent is a prolyl hydroxylase inhibitor.
3148. The method of item 3065 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3149. The method of item 3065 wherein the agent is a protein kinase B inhibitor.
3150. The method of item 3065 wherein the agent is a protein kinase C stimulant.
3151. The method of item 3065 wherein the agent is a purine nucleoside analogue.
3152. The method of item 3065 wherein the agent is a purinoreceptor P2X antagonist. 3153. The method of item 3065 wherein the agent is a Raf kinase inhibitor.
3154. The method of item 3065 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3155. The method of item 3065 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3156. The method of item 3065 wherein the agent is an SDF-1 antagonist.
3157. The method of item 3065 wherein the agent is a sheddase inhibitor.
3158. The method of item 3065 wherein the agent is an SRC inhibitor.
3159. The method of item 3065 wherein the agent is a stromelysin inhibitor.
I 3160. The method of item 3065 wherein the agent is an Syk kinase inhibitor.
3161. The method of item 3065 wherein the agent is a telomerase inhibitor.
3162. The method of item 3065 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No.
53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 3163. The method of item 3065 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3164. The method of item 3065 wherein the agent is a Toll receptor inhibitor.
3165. The method of item 3065 wherein the agent is a tubulin antagonist.
3166. The method of item 3065 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), 1GF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosyiate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab - and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3167. The method of item 3065 wherein the agent is a VEGF inhibitor.
3168. The method of item 3065 wherein the agent is a vitamin D receptor agonist.
3169. The method of item 3065 wherein the agent is ZD-6474 (an angiogenesis inhibitor). 3170. The method of item 3065 wherein the agent is AP-23573 (an mTOR inhibitor).
3171. The method of item 3065 wherein the agent is synthadotin (a tubulin antagonist).
3172. The method of item 3065 wherein the agent is S-0885 (a collagenase inhibitor).
3173. The method of item 3065 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3174. The method of item 3065 wherein the agent is ixabepilone (an epithilone).
3175. The method of item 3065 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3176. The method of item 3065 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3177. The method of item 3065 wherein the agent is ABT-518
(an angiogenesis inhibitor).
3178. The method of item 3065 wherein the agent is combretastatin (an angiogenesis inhibitor).
3179. The method of item 3065 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3180. The method of item 3065 wherein the agent is SB- 715992 (a kinesin antagonist).
3181. The method of item 3065 wherein the agent is temsirolimus (an mTOR inhibitor). 3182. The method of item 3065 wherein the agent is adalimumab (a TNFα antagonist).
3183. The method of item 3065, wherein the composition comprises a polymer.
3184. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
3185. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
3186. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
3187. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
3188. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
3189. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
3190. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
3191. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
3192. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 3193. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
3194. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
3195. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
3196. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
3197. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
3198. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
3199. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
3200. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
3201. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
3202. The method of item 3065, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
3203. The method of item 3065, wherein the composition further comprises a second pharmaceutically active agent. 3204. The method of item 3065, wherein the composition further comprises an anti-inflammatory agent.
3205. The method of item 3065, wherein the composition further comprises an agent that inhibits infection.
3206. The method of item 3065, wherein the composition further comprises an anthracycline.
3207. The method of item 3065, wherein the composition further comprises doxorubicin.
3208. The method of item 3065 wherein the composition further comprises mitoxantrone.
3209. The method of item 3065 wherein the composition further comprises a fluoropyrimidine.
3210. The method of item 3065, wherein the composition further comprises 5-fluorouracil (5-FU).
3211. The method of item 3065, wherein the composition further comprises a folic acid antagonist.
3212. The method of item 3065, wherein the composition further comprises methotrexate.
3213. The method of item 3065, wherein the composition further comprises a podophylotoxin.
3214. The method of item 3065, wherein the composition further comprises etoposide.
3215. The method of item 3065, wherein the composition further comprises camptothecin. 3216. The method of item 3065, wherein the composition further comprises a hydroxyurea.
3217. The method of item 3065, wherein the composition further comprises a platinum complex.
3218. The method of item 3065, wherein the composition further comprises cisplatin.
3219. The method of item 3065 wherein the composition further comprises an anti-thrombotic agent.
3220. The method of item 3065, wherein the composition further comprises a visualization agent.
3221. The method of item 3065, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3222. The method of item 3065, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
3223. The method of item 3065, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
3224. The method of item 3065, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
3225. The method of item 3065, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 3226. The method of item 3065, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
3227. The method of item 3065, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
3228. The method of item 3065 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
3229. The method of item 3065 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
3230. The method of item 3065 wherein the composition further comprises an inflammatory cytokine.
3231. The method of item 3065 wherein the composition further comprises an agent that stimulates cell proliferation.
3232. The method of item 3065 wherein the composition further comprises a polymeric carrier.
3233. The method of item 3065 wherein the composition is in the form of a gel, paste, or spray.
3234. The method of item 3065 wherein the sensor is partially constructed with the agent or the composition.
3235. The method of item 3065 wherein the sensor is impregnated with the agent or the composition. 3236. The method of item 3065, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
3237. The method of item 3065, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
3238. The method of item 3065 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
3239. The method of item 3065, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
3240. The method of item 3065 wherein the agent or the composition is located within pores or holes of the sensor.
3241. The method of item 3065 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
3242. The method of item 3065 wherein the sensor further comprises an echogenic material.
3243. The method of item 3065 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
3244. The method of item 3065 wherein the sensor is sterile.
3245. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
3246. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue. 3247. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
3248. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
3249. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
3250. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
3251. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
3252. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
3253. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
3254. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate. 3255. The method of item 3065 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
3256. The method of item 3065 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
3257. The method of item 3065 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
3258. The method of item 3065 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
3259. The method of item 3065 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
3260. The method of item 3065 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
3261. The method of item 3065 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
3262. The method of item 3065 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied. 3263. The method of item 3065 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
3264. The method of item 3065 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
3265. The method of item 3065 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
3266. The method of item 3065 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
3267. The method of item 3065, wherein the sensor further comprises a coating, and the coating is a uniform coating.
3268. The method of item 3065, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
3269. The method of item 3065, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
3270. The method of item 3065, wherein the sensor further comprises a coating, and the coating is a patterned coating.
3271. The method of item 3065, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less. 3272. The method of item 3065, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
3273. The method of item 3065, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
3274. The method of item 3065, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
3275. The method of item 3065, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
3276. The method of item 3065, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3277. The method of item 3065, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3278. The method of item 3065, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3279. The method of item 3065, wherein the sensor further comprises a coating, and the coating comprises a polymer.
3280. The method of item 3065, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition. 3281. The method of item 3065, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
3282. The method of item 3065 wherein the agent or the composition is affixed to the sensor.
3283. The method of item 3065 wherein the agent or the composition is covalently attached to the sensor.
3284. The method of item 3065 wherein the agent or the composition is non-covalently attached to the sensor.
3285. The method of item 3065 wherein the sensor comprises a coating that absorbs the agent or the composition.
3286. The method of item 3065 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
3287. The method of item 3065 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
3288. The method of item 3065 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
3289. The method of item 3065 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
3290. The method of item 3065 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
3291. The method of item 3065 wherein the sensor is linked to a pump. 3292. The method of item 3065 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
3293. The method of item 3065 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
3294. The method of item 3065 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
3295. The method of item 3065 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
3296. The method of item 3065 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
3297. The method of item 3065 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
3298. The method of item 3065 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
3299. The method of item 3065 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
3300. The method of any one of items 3065-3299 wherein the sensor is a blood or tissue glucose monitor. 3301. The method of any one of items 3065-3299 wherein the sensor is an electrolyte sensor.
3302. The method of any one of items 3065-3299 wherein the sensor is a blood constituent sensor.
3303. The method of any one of items 3065-3299 wherein the sensor is a temperature sensor.
3304. The method of any one of items 3065-3299 wherein the sensor is a pH sensor.
3305. The method of any one of items 3065-3299 wherein the sensor is an optical sensor.
3306. The method of any one of items 3065-3299 wherein the sensor is an amperometric sensor.
3307. The method of any one of items 3065-3299 wherein the sensor is a pressure sensor.
3308. The method of any one of items 3065-3299 wherein the sensor is a biosensor.
3309. The method of any one of items 3065-3299 wherein the sensor is a sensing transponder.
3310. The method of any one of items 3065-3299 wherein the sensor is a strain sensor.
3311. The method of any one of items 3065-3299 wherein the sensor is a magnetoresistive sensor.
3312. The method of any one of items 3065-3299 wherein the sensor is a cardiac sensor. 3313. The method of any one of items 3065-3299 wherein the sensor is a respiratory sensor.
3314. The method of any one of items 3065-3299 wherein the sensor is an auditory sensor.
3315. The method of any one of items 3065-3299 wherein the sensor is a metabolite sensor.
3316. The method of any one of items 3065-3299 wherein the sensor detects mechanical changes.
3317. The method of any one of items 3065-3299 wherein the sensor detects physical changes.
3318. The method of any one of items 3065-3299 wherein the sensor detects electrochemical changes.
3319. The method of any one of items 3065-3299 wherein the sensor detects magnetic changes.
3320. The method of any one of items 3065-3299 wherein the sensor detects acceleration changes.
3321. The method of any one of items 3065-3299 wherein the sensor detects ionizing radiation changes.
3322. The method of any one of items 3065-3299 wherein the sensor detects acoustic wave changes.
3323. The method of any one of items 3065-3299 wherein the sensor detects chemical changes.
3324. The method of any one of items 3065-3299 wherein the sensor detects drug concentration changes. 3325. The method of any one of items 3065-3299 wherein the sensor detects hormone changes.
3326. The method of any one of items 3065-3299 wherein the sensor detects barometric changes.
3327. A method for inhibiting scarring comprising placing a blood or tissue glucose monitor (i.e., a sensor) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
3328. The method of item 3327 wherein the agent is an adensosine A2A receptor antagonist.
3329. The method of item 3327 wherein the agent is an AKT inhibitor.
3330. The method of item 3327 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3331. The method of item 3327 wherein the agent is an alpha 4 integrin antagonist.
3332. The method of item 3327 wherein the agent is an alpha 7 nicotinic receptor agonist.
3333. The method of item 3327 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (Eli Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005
(GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3334. The method of item 3327 wherein the agent is an apoptosis antagonist.
3335. The method of item 3327 wherein the agent is an apoptosis activator.
3336. The method of item 3327 wherein the agent is a beta 1 integrin antagonist. 3337. The method of item 3327 wherein the agent is a beta tubulin inhibitor.
3338. The method of item 3327 wherein the agent is a blocker of enzyme production in Hepatitis C.
3339. The method of item 3327 wherein the agent is a Bruton's tyrosine kinase inhibitor.
3340. The method of item 3327 wherein the agent is a calcineurin inhibitor.
3341. The method of item 3327 wherein the agent is a caspase 3 inhibitor.
3342. The method of item 3327 wherein the agent is a CC chemokine receptor antagonist.
3343. The method of item 3327 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3344. The method of item 3327 wherein the agent is a cathepsin B inhibitor.
3345. The method of item 3327 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
3346. The method of item 3327 wherein the agent is a cathepsin L inhibitor.
3347. The method of item 3327 wherein the agent is a CD40 antagonist. 3348. The method of item 3327 wherein the agent is a chemokine receptor agonist.
3349. The method of item 3327 wherein the agent is a chymase inhibitor.
3350. The method of item 3327 wherein the agent is a collagenase antagonist.
3351. The method of item 3327 wherein the agent is a CXCR antagonist.
3352. The method of item 3327 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3353. The method of item 3327 wherein the agent is a cyclooxygenase 1 inhibitor.
3354. The method of item 3327 wherein the agent is a DHFR inhibitor.
3355. The method of item 3327 wherein the agent is a dual integrin inhibitor.
3356. The method of item 3327 wherein the agent is an elastase inhibitor. 3357. The method of item 3327 wherein the agent is an elongation factor-1 alpha inhibitor.
3358. The method of item 3327 wherein the agent is an endothelial growth factor antagonist.
3359. The method of item 3327 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3360. The method of item 3327 wherein the agent is an endotoxin antagonist.
3361. The method of item 3327 wherein the agent is an epothilone and tubulin binder.
3362. The method of item 3327 wherein the agent is an estrogen receptor antagonist.
3363. The method of item 3327 wherein the agent is an FGF inhibitor.
3364. The method of item 3327 wherein the agent is a farnexyl transferase inhibitor.
3365. The method of item 3327 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3366. The method of item 3327 wherein the agent is an FLT-3 kinase inhibitor.
3367. The method of item 3327 wherein the agent is an FGF receptor kinase inhibitor.
3368. The method of item 3327 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201
(Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3369. The method of item 3327 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
3370. The method of item 3327 wherein the agent is a histone deacetylase inhibitor.
3371. The method of item 3327 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 3372. The method of item 3327 wherein the agent is an ICAM inhibitor.
3373. The method of item 3327 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
3374. The method of item 3327 wherein the agent is an IL-2 inhibitor.
3375. The method of item 3327 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3376. The method of item 3327 wherein the agent is an IMPDH (inosine monophosphate). 3377. The method of item 3327 wherein the agent is an integrin antagonist.
3378. The method of item 3327 wherein the agent is an interleukin antagonist.
3379. The method of item 3327 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3380. The method of item 3327 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3381. The method of item 3327 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
3382. The method of item 3327 wherein the agent a JAK3 enzyme inhibitor.
3383. The method of item 3327 wherein the agent is a JNK inhibitor.
3384. The method of item 3327 wherein the agent is a kinase inhibitor.
3385. The method of item 3327 wherein the agent is kinesin antagonist.
3386. The method of item 3327 wherein the agent is a kinesin antagonist.
3387. The method of item 3327 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), FM 12 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) ' (Pfizer), and analogue or derivative thereof.
3388. The method of item 3327 wherein the agent is an MAP kinase inhibitor.
/ 3389. The method of item 3327 wherein the agent is a matrix metalloproteinase inhibitor.
3390. The method of item 3327 wherein the agent is an MCP-
CCR2 inhibitor.
3391. The method of item 3327 wherein the agent is an mTOR inhibitor.
3392. The method of item 3327 wherein the agent is an mTOR kinase inhibitor.
3393. The method of item 3327 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
3394. The method of item 3327 wherein the agent is an MIF inhibitor.
3395. The method of item 3327 wherein the agent is an MMP inhibitor.
3396. The method of item 3327 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
3397. The method of item 3327 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 3398. The method of item 3327 wherein the agent is a nitric oxide agonist.
3399. The method of item 3327 wherein the agent is an ornithine decarboxylase inhibitor.
3400. The method of item 3327 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3401. The method of item 3327 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3402. The method of item 3327 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3403. The method of item 3327 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3404. The method of item 3327 wherein the agent is a phosphatase inhibitor.
3405. The method of item 3327 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafenthne (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), 1BFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), trifiusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3406. The method of item 3327 wherein the agent is a PKC inhibitor.
3407. The method of item 3327 wherein the agent is a platelet activating factor antagonist.
3408. The method of item 3327 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3409. The method of item 3327 wherein the agent is a prolyl hydroxylase inhibitor.
3410. The method of item 3327 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3411. The method of item 3327 wherein the agent is a protein kinase B inhibitor.
3412. The method of item 3327 wherein the agent is a protein kinase C stimulant.
3413. The method of item 3327 wherein the agent is a purine nucleoside analogue.
3414. The method of item 3327 wherein the agent is a purinoreceptor P2X antagonist.
3415. The method of item 3327 wherein the agent is a Raf kinase inhibitor. 3416. The method of item 3327 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3417. The method of item 3327 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3418. The method of item 3327 wherein the agent is an SDF-1 antagonist.
3419. The method of item 3327 wherein the agent is a sheddase inhibitor.
3420. The method of item 3327 wherein the agent is an SRC inhibitor.
3421. The method of item 3327 wherein the agent is a stromelysin inhibitor.
3422. The method of item 3327 wherein the agent is an Syk kinase inhibitor.
3423. The method of item 3327 wherein the agent is a telomerase inhibitor.
3424. The method of item 3327 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3425. The method of item 3327 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Won Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3426. The method of item 3327 wherein the agent is a Toll receptor inhibitor.
3427. The method of item 3327 wherein the agent is a tubulin antagonist.
3428. The method of item 3327 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHlR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), Iapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NlH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3429. The method of item 3327 wherein the agent is a VEGF inhibitor.
3430. The method of item 3327 wherein the agent is a vitamin D receptor agonist.
3431. The method of item 3327 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
3432. The method of item 3327 wherein the agent is AP-23573 (an mTOR inhibitor). 3433. The method of item 3327 wherein the agent is synthadotin (a tubulin antagonist).
3434. The method of item 3327 wherein the agent is S-0885 (a collagenase inhibitor).
3435. The method of item 3327 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
3436. The method of item 3327 wherein the agent is ixabepilone (an epithilone).
3437. The method of item 3327 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3438. The method of item 3327 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3439. The method of item 3327 wherein the agent is ABT-518 (an angiogenesis inhibitor).
3440. The method of item 3327 wherein the agent is combretastatin (an angiogenesis inhibitor).
3441. The method of item 3327 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3442. The method of item 3327 wherein the agent is SB- 715992 (a kinesin antagonist).
3443. The method of item 3327 wherein the agent is temsirolimus (an mTOR inhibitor).
3444. The method of item 3327 wherein the agent is adalimumab (a TNFα antagonist). 3445. The method of item 3327, wherein the composition comprises a polymer.
3446. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
3447. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
3448. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
3449. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
3450. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
3451. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
3452. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
3453. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
3454. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 3455. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
3456. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
3457. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
3458. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
3459. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
3460. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
3461. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
3462. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
3463. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
3464. The method of item 3327, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
3465. The method of item 3327, wherein the composition further comprises a second pharmaceutically active agent. 3466. The method of item 3327, wherein the composition further comprises an anti-inflammatory agent.
3467. The method of item 3327, wherein the composition further comprises an agent that inhibits infection.
3468. The method of item 3327, wherein the composition further comprises an anthracycline.
3469. The method of item 3327, wherein the composition further comprises doxorubicin.
3470. The method of item 3327 wherein the composition further comprises mitoxantrone.
3471. The method of item 3327 wherein the composition further comprises a fluoropyrimidine.
3472. The method of item 3327, wherein the composition further comprises 5-fluorouracil (5-FU).
3473. The method of item 3327, wherein the composition further comprises a folic acid antagonist.
3474. The method of item 3327, wherein the composition further comprises methotrexate.
3475. The method of item 3327, wherein the composition further comprises a podophylotoxin.
3476. The method of item 3327, wherein the composition further comprises etoposide.
3477. The method of item 3327, wherein the composition further comprises camptothecin. 3478. The method of item 3327, wherein the composition further comprises a hydroxyurea.
3479. The method of item 3327, wherein the composition further comprises a platinum complex.
3480. The method of item 3327, wherein the composition further comprises cisplatin.
3481. The method of item 3327 wherein the composition further comprises an anti-thrombotic agent.
3482. The method of item 3327, wherein the composition further comprises a visualization agent.
3483. The method of item 3327, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3484. The method of item 3327, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
3485. The method of item 3327, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
3486. The method of item 3327, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
3487. The method of item 3327, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 3488. The method of item 3327, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
3489. The method of item 3327, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
3490. The method of item 3327 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
3491. The method of item 3327 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
3492. The method of item 3327 wherein the composition further comprises an inflammatory cytokine.
3493. The method of item 3327 wherein the composition further comprises an agent that stimulates cell proliferation.
3494. The method of item 3327 wherein the composition further comprises a polymeric carrier.
3495. The method of item 3327 wherein the composition is in the form of a gel, paste, or spray.
3496. The method of item 3327 wherein the sensor is partially constructed with the agent or the composition.
3497. The method of item 3327 wherein the sensor is impregnated with the agent or the composition. 3498. The method of item 3327, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
3499. The method of item 3327, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
3500. The method of item 3327 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
3501. The method of item 3327, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
3502. The method of item 3327 wherein the agent or the composition is located within pores or holes of the sensor.
3503. The method of item 3327 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
3504. The method of item 3327 wherein the sensor further comprises an echogenic material.
3505. The method of item 3327 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
3506. The method of item 3327 wherein the sensor is sterile.
3507. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
3508. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue. 3509. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
3510. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
3511. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
3512. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
3513. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
3514. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
3515. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
3516. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate. 3517. The method of item 3327 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
3518. The method of item 3327 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
3519. The method of item 3327 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
3520. The method of item 3327 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
3521. The method of item 3327 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
3522. The method of item 3327 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
3523. The method of item 3327 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
3524. The method of item 3327 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied. 3525. The method of item 3327 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
3526. The method of item 3327 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
3527. The method of item 3327 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
3528. The method of item 3327 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
3529. The method of item 3327, wherein the sensor further comprises a coating, and the coating is a uniform coating.
3530. The method of item 3327, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
3531. The method of item 3327, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
3532. The method of item 3327, wherein the sensor further comprises a coating, and the coating is a patterned coating.
3533. The method of item 3327, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less. 3534. The method of item 3327, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
3535. The method of item 3327, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
3536. The method of item 3327, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
3537. The method of item 3327, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3538. The method of item 3327, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3539. The method of item 3327, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3540. The method of item 3327, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3541. The method of item 3327, wherein the sensor further comprises a coating, and the coating comprises a polymer.
3542. The method of item 3327, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition. 3543. The method of item 3327, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
3544. The method of item 3327 wherein the agent or the composition is affixed to the sensor.
3545. The method of item 3327 wherein the agent or the composition is covalently attached to the sensor.
3546. The method of item 3327 wherein the agent or the composition is non-covalently attached to the sensor.
3547. The method of item 3327 wherein the sensor comprises a coating that absorbs the agent or the composition.
3548. The method of item 3327 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
3549. The method of item 3327 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
3550. The method of item 3327 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
3551. The method of item 3327 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
3552. The method of item 3327 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
3553. The method of item 3327 wherein the sensor is linked to a pump. 3554. The method of item 3327 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
3555. The method of item 3327 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
3556. The method of item 3327 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
3557. The method of item 3327 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
3558. The method of item 3327 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
3559. The method of item 3327 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
3560. The method of item 3327 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
3561. The method of item 3327 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
3562. The method of any one of items 3327-3561 wherein the sensor is deliverable to the vascular system transluminal^ using a catheter on a stent platform. 3563. The method of any one of items 3327-3561 wherein the sensor is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
3564. The method of any one of items 3327-3561 wherein the sensor is an electrode composed of an analyte responsive enzyme.
3565. The method of any one of items 3327-3561 wherein the sensor is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
3566. The method of any one of items 3327-3561 wherein the sensor is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin.
3567. A method for inhibiting scarring comprising placing a pressure or stress sensor and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
3568. The method of item 3567 wherein the agent is an adensosine A2A receptor antagonist.
3569. The method of item 3567 wherein the agent is an AKT inhibitor.
3570. The method of item 3567 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3571. The method of item 3567 wherein the agent is an alpha 4 integrin antagonist. 3572. The method of item 3567 wherein the agent is an alpha 7 nicotinic receptor agonist.
3573. The method of item 3567 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BϊoTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEΞAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3574. The method of item 3567 wherein the agent is an apoptosis antagonist.
3575. The method of item 3567 wherein the agent is an apoptosis activator.
3576. The method of item 3567 wherein the agent is a beta 1 integrin antagonist.
3577. The method of item 3567 wherein the agent is a beta tubulin inhibitor.
3578. The method of item 3567 wherein the agent is a blocker of enzyme production in Hepatitis C.
3579. The method of item 3567 wherein the agent is a Bruton's tyrosine kinase inhibitor.
3580. The method of item 3567 wherein the agent is a calcineurin inhibitor.
3581. The method of item 3567 wherein the agent is a caspase 3 inhibitor.
, 3582. The method of item 3567 wherein the agent is a CC chemokine receptor antagonist.
3583. The method of item 3567 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3584. The method of item 3567 wherein the agent is a cathepsin B inhibitor. 3585. The method of item 3567 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
3586. The method of item 3567 wherein the agent is a cathepsin L inhibitor.
3587. The method of item 3567 wherein the agent is a CD40 antagonist.
3588. The method of item 3567 wherein the agent is a chemokine receptor agonist.
3589. The method of item 3567 wherein the agent is a chymase inhibitor.
3590. The method of item 3567 wherein the agent is a collagenase antagonist.
3591. The method of item 3567 wherein the agent is a CXCR antagonist.
3592. The method of item 3567 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. ooao. i πe meiπoα oτ iiem ύoof wnerein me agent is a cyclooxygenase 1 inhibitor.
3594. The method of item 3567 wherein the agent is a DHFR inhibitor.
3595. The method of item 3567 wherein the agent is a dual integrin inhibitor.
3596. The method of item 3567 wherein the agent is an elastase inhibitor.
3597. The method of item 3567 wherein the agent is an elongation factor- 1 alpha inhibitor.
3598. The method of item 3567 wherein the agent is an endothelial growth factor antagonist.
3599. The method of item 3567 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3600. The method of item 3567 wherein the agent is an endotoxin antagonist.
3601. The method of item 3567 wherein the agent is an epothilone and tubulin binder. 3602. The method of item 3567 wherein the agent is an estrogen receptor antagonist.
3603. The method of item 3567 wherein the agent is an FGF inhibitor.
3604. The method of item 3567 wherein the agent is a farnexyl transferase inhibitor.
3605. The method of item 3567 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme),
Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3606. The method of item 3567 wherein the agent is an FLT-3 kinase inhibitor.
3607. The method of item 3567 wherein the agent is an FGF receptor kinase inhibitor.
3608. The method of item 3567 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11 -9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3609. The method of item 3567 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XlV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
3610. The method of item 3567 wherein the agent is a histone deacetylase inhibitor.
3611. The method of item 3567 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
3612. The method of item 3567 wherein the agent is an ICAM inhibitor.
3613. The method of item 3567 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
3614. The method of item 3567 wherein the agent is an IL-2 inhibitor.
3615. The method of item 3567 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3616. The method of item 3567 wherein the agent is an IMPDH (inosine monophosphate).
3617. The method of item 3567 wherein the agent is an integrin antagonist.
3618. The method of item 3567 wherein the agent is an interleukin antagonist.
3619. The method of item 3567 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3620. The method of item 3567 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3621. The method of item 3567 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
3622. The method of item 3567 wherein the agent a JAK3 enzyme inhibitor.
3623. The method of item 3567 wherein the agent is a JNK inhibitor.
3624. The method of item 3567 wherein the agent is a kinase inhibitor. 3625. The method of item 3567 wherein the agent is kinesin antagonist.
3626. The method of item 3567 wherein the agent is a kinesin antagonist.
3627. The method of item 3567 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3628. The method of item 3567 wherein the agent is an MAP kinase inhibitor.
3629. The method of item 3567 wherein the agent is a matrix metalloproteinase inhibitor.
3630. The method of item 3567 wherein the agent is an MCP-
CCR2 inhibitor.
3631. The method of item 3567 wherein the agent is an mTOR inhibitor. 3632. The method of item 3567 wherein the agent is an mTOR kinase inhibitor.
3633. The method of item 3567 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), GenexoI-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
3634. The method of item 3567 wherein the agent is an MIF inhibitor.
3635. The method of item 3567 wherein the agent is an MMP inhibitor.
3636. The method of item 3567 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 3637. The method of item 3567 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3638. The method of item 3567 wherein the agent is a nitric oxide agonist.
3639. The method of item 3567 wherein the agent is an ornithine decarboxylase inhibitor.
3640. The method of item 3567 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3641. The method of item 3567 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3642. The method of item 3567 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3643. The method of item 3567 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3644. The method of item 3567 wherein the agent is a phosphatase inhibitor.
3645. The method of item 3567 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafenthne (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, 1BFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3646. The method of item 3567 wherein the agent is a PKC inhibitor.
3647. The method of item 3567 wherein the agent is a platelet activating factor antagonist.
3648. The method of item 3567 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3649. The method of item 3567 wherein the agent is a prolyl hydroxylase inhibitor.
3650. The method of item 3567 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3651. The method of item 3567 wherein the agent is a protein kinase B inhibitor. 3652. The method of item 3567 wherein the agent is a protein kinase C stimulant.
3653. The method of item 3567 wherein the agent is a purine nucleoside analogue.
3654. The method of item 3567 wherein the agent is a purinoreceptor P2X antagonist.
3655. The method of item 3567 wherein the agent is a Raf kinase inhibitor.
3656. The method of item 3567 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3657. The method of item 3567 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3658. The method of item 3567 wherein the agent is an SDF-1 antagonist.
3659. The method of item 3567 wherein the agent is a sheddase inhibitor.
3660. The method of item 3567 wherein the agent is an SRC inhibitor.
3661. The method of item 3567 wherein the agent is a stromelysin inhibitor.
3662. The method of item 3567 wherein the agent is an Syk kinase inhibitor.
3663. The method of item 3567 wherein the agent is a telomerase inhibitor. 3664. The method of item 3567 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG)1 TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3665. The method of item 3567 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3666. The method of item 3567 wherein the agent is a Toll receptor inhibitor.
3667. The method of item 3567 wherein the agent is a tubulin antagonist.
3668. The method of item 3567 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 3669. The method of item 3567 wherein the agent is a VEGF inhibitor.
3670. The method of item 3567 wherein the agent is a vitamin D receptor agonist.
3671. The method of item 3567 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
3672. The method of item 3567 wherein the agent is AP-23573 (an mTOR inhibitor).
3673. The method of item 3567 wherein the agent is synthadotin (a tubulin antagonist).
3674. The method of item 3567 wherein the agent is S-0885 (a collagenase inhibitor).
3675. The method of item 3567 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3676. The method of item 3567 wherein the agent is ixabepilone (an epithilone).
3677. The method of item 3567 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3678. The method of item 3567 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3679. The method of item 3567 wherein the agent is ABT-518 (an angiogenesis inhibitor).
3680. The method of item 3567 wherein the agent is combretastatin (an angiogenesis inhibitor). 3681. The method of item 3567 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3682. The method of item 3567 wherein the agent is SB- 715992 (a kinesin antagonist).
3683. The method of item 3567 wherein the agent is temsirolimus (an mTOR inhibitor).
3684. The method of item 3567 wherein the agent is adalimumab (a TNFα antagonist).
3685. The method of item 3567, wherein the composition comprises a polymer.
3686. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
3687. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
3688. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
3689. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
3690. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
3691. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer. 3692. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
3693. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
3694. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
3695. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
3696. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
3697. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
3698. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
3699. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
3700. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
3701. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer. 3702. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
3703. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
3704. The method of item 3567, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
3705. The method of item 3567, wherein the composition further comprises a second pharmaceutically active agent.
3706. The method of item 3567, wherein the composition further comprises an anti-inflammatory agent.
3707. The method of item 3567, wherein the composition further comprises an agent that inhibits infection.
3708. The method of item 3567, wherein the composition further comprises an anthracycline.
3709. The method of item 3567, wherein the composition further comprises doxorubicin.
3710. The method of item 3567 wherein the composition further comprises mitoxantrone.
3711. The method of item 3567 wherein the composition further comprises a fluoropyrimidine.
3712. The method of item 3567, wherein the composition further comprises 5-fluorouracil (5-FU).
3713. The method of item 3567, wherein the composition further comprises a folic acid antagonist. 3714. The method of item 3567, wherein the composition further comprises methotrexate.
3715. The method of item 3567, wherein the composition further comprises a podophylotoxin.
3716. The method of item 3567, wherein the composition further comprises etoposide.
3717. The method of item 3567, wherein the composition further comprises camptothecin.
3718. The method of item 3567, wherein the composition further comprises a hydroxyurea.
3719. The method of item 3567, wherein the composition further comprises a platinum complex.
3720. The method of item 3567, wherein the composition further comprises cisplatin.
3721. The method of item 3567 wherein the composition further comprises an anti-thrombotic agent.
3722. The method of item 3567, wherein the composition further comprises a visualization agent.
3723. The method of item 3567, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3724. The method of item 3567, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium. 3725. The method of item 3567, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
3726. The method of item 3567, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
3727. The method of item 3567, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
3728: The method of item 3567, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
3729. The method of item 3567, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
3730. The method of item 3567 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
3731. The method of item 3567 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
3732. The method of item 3567 wherein the composition further comprises an inflammatory cytokine.
3733. The method of item 3567 wherein the composition further comprises an agent that stimulates cell proliferation. 3734. The method of item 3567 wherein the composition further comprises a polymeric carrier.
3735. The method of item 3567 wherein the composition is in the form of a gel, paste, or spray.
3736. The method of item 3567 wherein the sensor is partially constructed with the agent or the composition.
3737. The method of item 3567 wherein the sensor is impregnated with the agent or the composition.
3738. The method of item 3567, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
3739. The method of item 3567, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
3740. The method of item 3567 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
3741. The method of item 3567, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
3742. The method of item 3567 wherein the agent or the composition is located within pores or holes of the sensor.
3743. The method of item 3567 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
3744. The method of item 3567 wherein the sensor further comprises an echogenic material.
3745. The method of item 3567 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating. 3746. The method of item 3567 wherein the sensor is sterile.
3747. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
3748. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
3749. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
3750. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
3751. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
3752. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
3753. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
3754. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days. 3755. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
3756. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
3757. The method of item 3567 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
3758. The method of item 3567 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
3759. The method of item 3567 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
3760. The method of item 3567 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
3761. The method of item 3567 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
3762. The method of item 3567 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
3763. The method of item 3567 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied. 3764. The method of item 3567 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
3765. The method of item 3567 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
3766. The method of item 3567 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
3767. The method of item 3567 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
3768. The method of item 3567 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
3769. The method of item 3567, wherein the sensor further comprises a coating, and the coating is a uniform coating.
3770. The method of item 3567, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
3771. The method of item 3567, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
3772. The method of item 3567, wherein the sensor further comprises a coating, and the coating is a patterned coating. 3773. The method of item 3567, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
3774. The method of item 3567, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
3775. The method of item 3567, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
3776. The method of item 3567, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
3777. The method of item 3567, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3778. The method of item 3567, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3779. The method of item 3567, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3780. The method of item 3567, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3781. The method of item 3567, wherein the sensor further comprises a coating, and the coating comprises a polymer. 3782. The method of item 3567, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
3783. The method of item 3567, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
3784. The method of item 3567 wherein the agent or the composition is affixed to the sensor.
3785. The method of item 3567 wherein the agent or the composition is covalently attached to the sensor.
3786. The method of item 3567 wherein the agent or the composition is non-covalently attached to the sensor.
3787. The method of item 3567 wherein the sensor comprises a coating that absorbs the agent or the composition.
3788. The method of item 3567 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
3789. The method of item 3567 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
3790. The method of item 3567 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
3791. The method of item 3567 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
3792. The method of item 3567 wherein the sensor is completely covered with a mesh that contains the agent or the composition. 3793. The method of item 3567 wherein the sensor is linked to a pump.
3794. The method of item 3567 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
3795. The method of item 3567 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
3796. The method of item 3567 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
3797. The method of item 3567 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
3798. The method of item 3567 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
3799. The method of item 3567 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
3800. The method of item 3567 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
3801. The method of item 3567 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor. 3802. The method of any one of items 3567-3801 wherein the sensor monitors blood pressure.
3803. The method of any one of items 3567-3801 wherein the sensor monitors fluid flow.
3804. The method of any one of items 3567-3801 wherein the sensor monitors pressure within an aneurysm sac.
3805. The method of any one of items 3567-3801 wherein the sensor monitors intracranial pressure.
3806. The method of any one of items 3567-3801 wherein the sensor monitors mechanical pressure associated with a bone fracture.
3807. The method of any one of items 3567-3801 wherein the sensor monitors barometric pressure.
3808. The method of any one of items 3567-3801 wherein the sensor monitors eye tremors.
3809. The method of any one of items 3567-3801 wherein the sensor monitors the depth of a corneal implant.
3810. The method of any one of items 3567-3801 wherein the sensor monitors intraocular pressure.
3811. The method of any one of items 3567-3801 wherein the sensor is a passive sensor with an inductor-capacitor circuit.
3812. The method of any one of items 3567-3801 wherein the sensor is a self-powered strain sensing system.
3813. The method of any one of items 3567-3801 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage. 3814. A method for inhibiting scarring comprising placing a cardiac sensor and an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
3815. The method of item 3814 wherein the agent is an adensosine A2A receptor antagonist.
3816. The method of item 3814 wherein the agent is an AKT inhibitor.
3817. The method of item 3814 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3818. The method of item 3814 wherein the agent is an alpha 4 integrin antagonist.
3819. The method of item 3814 wherein the agent is an alpha 7 nicotinic receptor agonist.
3820. The method of item 3814 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), H1F-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB)1 JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3821. The method of item 3814 wherein the agent is an apoptosis antagonist.
3822. The method of item 3814 wherein the agent is an apoptosis activator.
3823. The method of item 3814 wherein the agent is a beta 1 integrin antagonist.
3824. The method of item 3814 wherein the agent is a beta tubulin inhibitor.
3825. The method of item 3814 wherein the agent is a blocker of enzyme production in Hepatitis C.
3826. The method of item 3814 wherein the agent is a Bruton's tyrosine kinase inhibitor. 3827. The method of item 3814 wherein the agent is a calcineurin inhibitor.
3828. The method of item 3814 wherein the agent is a caspase 3 inhibitor.
3829. The method of item 3814 wherein the agent is a CC chemokine receptor antagonist.
3830. The method of item 3814 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3831. The method of item 3814 wherein the agent is a cathepsin B inhibitor.
3832. The method of item 3814 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
3833. The method of item 3814 wherein the agent is a cathepsin L inhibitor.
3834. The method of item 3814 wherein the agent is a CD40 antagonist.
3835. The method of item 3814 wherein the agent is a chemokine receptor agonist.
3836. The method of item 3814 wherein the agent is a chymase inhibitor.
3837. The method of item 3814 wherein the agent is a collagenase antagonist. 3838. The method of item 3814 wherein the agent is a CXCR antagonist.
3839. The method of item 3814 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3840. The method of item 3814 wherein the agent is a cyclooxygenase 1 inhibitor.
3841. The method of item 3814 wherein the agent is a DHFR inhibitor.
3842. The method of item 3814 wherein the agent is a dual ϊntegrin inhibitor.
3843. The method of item 3814 wherein the agent is an elastase inhibitor.
3844. The method of item 3814 wherein the agent is an elongation factor-1 alpha inhibitor.
3845. The method of item 3814 wherein the agent is an endothelial growth factor antagonist.
3846. The method of item 3814 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI
Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3847. The method of item 3814 wherein the agent is an endotoxin antagonist.
3848. The method of item 3814 wherein the agent is an epothilone and tubulin binder.
3849. The method of item 3814 wherein the agent is an estrogen receptor antagonist.
3850. The method of item 3814 wherein the agent is an FGF inhibitor.
3851. The method of item 3814 wherein the agent is a famexyl transferase inhibitor.
3852. The method of item 3814 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3853. The method of item 3814 wherein the agent is an FLT-3 kinase inhibitor. 3854. The method of item 3814 wherein the agent is an FGF receptor kinase inhibitor.
3855. The method of item 3814 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3856. The method of item 3814 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005
(Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
3857. The method of item 3814 wherein the agent is a histone deacetylase inhibitor.
3858. The method of item 3814 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
3859. The method of item 3814 wherein the agent is an ICAM inhibitor.
3860. The method of item 3814 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 3861. The method of item 3814 wherein the agent is an IL-2 inhibitor.
3862. The method of item 3814 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No.457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3863. The method of item 3814 wherein the agent is an IMPDH (inosine monophosphate).
3864. The method of item 3814 wherein the agent is an integrin antagonist.
3865. The method of item 3814 wherein the agent is an interleukin antagonist.
3866. The method of item 3814 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 3867. The method of item 3814 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3868. The method of item 3814 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
3869. The method of item 3814 wherein the agent a JAK3 enzyme inhibitor.
3870. The method of item 3814 wherein the agent is a JNK inhibitor.
3871. The method of item 3814 wherein the agent is a kinase inhibitor.
3872. The method of item 3814 wherein the agent is kinesin antagonist.
3873. The method of item 3814 wherein the agent is a kinesin antagonist.
3874. The method of item 3814 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucothene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3875. The method of item 3814 wherein the agent is an MAP kinase inhibitor.
3876. The method of item 3814 wherein the agent is a matrix metalloproteinase inhibitor.
3877. The method of item 3814 wherein the agent is an MCP- CCR2 inhibitor.
3878. The method of item 3814 wherein the agent is an mTOR inhibitor.
3879. The method of item 3814 wherein the agent is an mTOR kinase inhibitor.
3880. The method of item 3814 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
3881. The method of item 3814 wherein the agent is an MIF inhibitor. 3882. The method of item 3814 wherein the agent is an MMP inhibitor.
3883. The method of item 3814 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
3884. The method of item 3814 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3885. The method of item 3814 wherein the agent is a nitric oxide agonist.
3886. The method of item 3814 wherein the agent is an ornithine decarboxylase inhibitor.
3887. The method of item 3814 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3888. The method of item 3814 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3889. The method of item 3814 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-
860 (UCB)1 E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3890. The method of item 3814 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET
(GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3891. The method of item 3814 wherein the agent is a phosphatase inhibitor.
3892. The method of item 3814 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 3893. The method of item 3814 wherein the agent is a PKC inhibitor.
3894. The method of item 3814 wherein the agent is a platelet activating factor antagonist.
3895. The method of item 3814 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3896. The method of item 3814 wherein the agent is a prolyl hydroxylase inhibitor.
3897. The method of item 3814 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3898. The method of item 3814 wherein the agent is a protein kinase B inhibitor.
3899. The method of item 3814 wherein the agent is a protein kinase C stimulant.
3900. The method of item 3814 wherein the agent is a purine nucleoside analogue.
3901. The method of item 3814 wherein the agent is a purinoreceptor P2X antagonist.
3902. The method of item 3814 wherein the agent is a Raf kinase inhibitor.
3903. The method of item 3814 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3904. The method of item 3814 wherein the agent is a ribonucleoside triphosphate reductase inhibitor. 3905. The method of item 3814 wherein the agent is an SDF-1 antagonist.
3906. The method of item 3814 wherein the agent is a sheddase inhibitor.
3907. The method of item 3814 wherein the agent is an SRC inhibitor.
3908. The method of item 3814 wherein the agent is a stromelysin inhibitor.
3909. The method of item 3814 wherein the agent is an Syk kinase inhibitor.
3910. The method of item 3814 wherein the agent is a telomerase inhibitor.
3911. The method of item 3814 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3912. The method of item 3814 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 3913. The method of item 3814 wherein the agent is a Toll receptor inhibitor.
3914. The method of item 3814 wherein the agent is a tubulin antagonist.
3915. The method of item 3814 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis
Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3916. The method of item 3814 wherein the agent is a VEGF inhibitor.
3917. The method of item 3814 wherein the agent is a vitamin D receptor agonist.
3918. The method of item 3814 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
3919. The method of item 3814 wherein the agent is AP-23573 (an mTOR inhibitor).
3920. The method of item 3814 wherein the agent is synthadotin (a tubulin antagonist).
3921. The method of item 3814 wherein the agent is S-0885 (a collagenase inhibitor). 3922. The method of item 3814 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3923. The method of item 3814 wherein the agent is ixabepilone (an epithilone).
3924. The method of item 3814 wherein the agent is IDN-5390
(an angiogenesis inhibitor).
3925. The method of item 3814 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3926. The method of item 3814 wherein the agent is ABT-518 (an angiogenesis inhibitor).
3927. The method of item 3814 wherein the agent is combretastatin (an angiogenesis inhibitor).
3928. The method of item 3814 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3929. The method of item 3814 wherein the agent is SB-
715992 (a kinesin antagonist).
3930. The method of item 3814 wherein the agent is temsirolimus (an mTOR inhibitor).
3931. The method of item 3814 wherein the agent is adalimumab (a TNFα antagonist).
3932. The method of item 3814, wherein the composition comprises a polymer.
3933. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 3934. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
3935. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
3936. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
3937. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
3938. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
3939. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
3940. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
3941. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
3942. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
3943. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer. 3944. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
3945. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
3946. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
3947. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
3948. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
3949. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
3950. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
3951. The method of item 3814, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
3952. The method of item 3814, wherein the composition further comprises a second pharmaceutically active agent.
3953. The method of item 3814, wherein the composition further comprises an anti-inflammatory agent.
3954. The method of item 3814, wherein the composition further comprises an agent that inhibits infection. 3955. The method of item 3814, wherein the composition further comprises an anthracycline.
3956. The method of item 3814, wherein the composition further comprises doxorubicin.
3957. The method of item 3814 wherein the composition further comprises mitoxantrone.
3958. The method of item 3814 wherein the composition further comprises a fluoropyrimidine.
3959. The method of item 3814, wherein the composition further comprises 5-fluorouracil (5-FU).
3960. The method of item 3814, wherein the composition further comprises a folic acid antagonist.
3961. The method of item 3814, wherein the composition further comprises methotrexate.
3962. The method of item 3814, wherein the composition further comprises a podophylotoxin.
3963. The method of item 3814, wherein the composition further comprises etoposide.
3964. The method of item 3814, wherein the composition further comprises camptothecin.
3965. The method of item 3814, wherein the composition further comprises a hydroxyurea.
3966. The method of item 3814, wherein the composition further comprises a platinum complex. 3967. The method of item 3814, wherein the composition further comprises cisplatin.
3968. The method of item 3814 wherein the composition further comprises an anti-thrombotic agent.
3969. The method of item 3814, wherein the composition further comprises a visualization agent.
3970. The method of item 3814, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3971. The method of item 3814, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
3972. The method of item 3814, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
3973. The method of item 3814, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
3974. The method of item 3814, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
3975. The method of item 3814, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound. 3976. The method of item 3814, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
3977. The method of item 3814 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
3978. The method of item 3814 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
3979. The method of item 3814 wherein the composition further comprises an inflammatory cytokine.
3980. The method of item 3814 wherein the composition further comprises an agent that stimulates cell proliferation.
3981. The method of item 3814 wherein the composition further comprises a polymeric carrier.
3982. The method of item 3814 wherein the composition is in the form of a gel, paste, or spray.
3983. The method of item 3814 wherein the sensor is partially constructed with the agent or the composition.
3984. The method of item 3814 wherein the sensor is impregnated with the agent or the composition.
3985. The method of item 3814, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
3986. The method of item 3814, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor. 3987. The method of item 3814 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
3988. The method of item 3814, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
3989. The method of item 3814 wherein the agent or the composition is located within pores or holes of the sensor.
3990. The method of item 3814 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
3991. The method of item 3814 wherein the sensor further comprises an echogenic material.
3992. The method of item 3814 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
3993. The method of item 3814 wherein the sensor is sterile.
3994. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
3995. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
3996. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
3997. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue. 3998. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
3999. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
4000. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
4001. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
4002. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
4003. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
4004. The method of item 3814 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
4005. The method of item 3814 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent. 4006. The method of item 3814 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
4007. The method of item 3814 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
4008. The method of item 3814 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
4009. The method of item 3814 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
4010. The method of item 3814 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
4011. The method of item 3814 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
4012. The method of item 3814 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
4013. The method of item 3814 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied. 4014. The method of item 3814 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
4015. The method of item 3814 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
4016. The method of item 3814, wherein the sensor further comprises a coating, and the coating is a uniform coating.
4017. The method of item 3814, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
4018. The method of item 3814, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
4019. The method of item 3814, wherein the sensor further comprises a coating, and the coating is a patterned coating.
4020. The method of item 3814, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
4021. The method of item 3814, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
4022. The method of item 3814, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
4023. The method of item 3814, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year. 4024. The method of item 3814, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4025. The method of item 3814, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4026. The method of item 3814, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4027. The method of item 3814, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4028. The method of item 3814, wherein the sensor further comprises a coating, and the coating comprises a polymer.
4029. The method of item 3814, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
4030. The method of item 3814, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
4031. The method of item 3814 wherein the agent or the composition is affixed to the sensor.
4032. The method of item 3814 wherein the agent or the composition is covalently attached to the sensor. 4033. The method of item 3814 wherein the agent or the composition is non-covalently attached to the sensor.
4034. The method of item 3814 wherein the sensor comprises a coating that absorbs the agent or the composition.
4035. The method of item 3814 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
4036. The method of item 3814 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
4037. The method of item 3814 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
4038. The method of item 3814 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
4039. The method of item 3814 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
4040. The method of item 3814 wherein the sensor is linked to a pump.
4041. The method of item 3814 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
4042. The method of item 3814 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
4043. The method of item 3814 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host. 4044. The method of item 3814 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
4045. The method of item 3814 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
4046. The method of item 3814 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
4047. The method of item 3814 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
4048. The method of item 3814 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
4049. The method of any one of items 3814-4048 wherein the sensor monitors cardiac output.
4050. The method of any one of items 3814-4048 wherein the sensor monitors ejection fraction.
4051. The method of any one of items 3814-4048 wherein the sensor monitors blood pressure in a heart chamber.
4052. The method of any one of items 3814-4048 wherein the sensor monitors ventricular wall motions.
4053. The method of any one of items 3814-4048 wherein the sensor monitors blood flow to a transplanted organ. 4054. The method of any one of items 3814-4048 wherein the sensor monitors heart rate.
4055. A method for inhibiting scarring comprising placing a respiratory sensor and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
4056. The method of item 4055 wherein the agent is an adensosine A2A receptor antagonist.
4057. The method of item 4055 wherein the agent is an AKT inhibitor.
4058. The method of item 4055 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4059. The method of item 4055 wherein the agent is an alpha 4 integrin antagonist.
4060. The method of item 4055 wherein the agent is an alpha 7 nicotinic receptor agonist.
4061. The method of item 4055 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4062. The method of item 4055 wherein the agent is an apoptosis antagonist.
4063. The method of item 4055 wherein the agent is an apoptosis activator.
4064. The method of item 4055 wherein the agent is a beta 1 integrin antagonist.
4065. The method of item 4055 wherein the agent is a beta tubulin inhibitor. 4066. The method of item 4055 wherein the agent is a blocker of enzyme production in Hepatitis C.
4067. The method of item 4055 wherein the agent is a Bruton's tyrosine kinase inhibitor.
4068. The method of item 4055 wherein the agent is a calcineurin inhibitor.
4069. The method of item 4055 wherein the agent is a caspase 3 inhibitor.
4070. The method of item 4055 wherein the agent is a CC chemokine receptor antagonist.
4071. The method of item 4055 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4072. The method of item 4055 wherein the agent is a cathepsin B inhibitor.
4073. The method of item 4055 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
4074. The method of item 4055 wherein the agent is a cathepsin L inhibitor.
4075. The method of item 4055 wherein the agent is a CD40 antagonist.
4076. The method of item 4055 wherein the agent is a chemokine receptor agonist. 4077. The method of item 4055 wherein the agent is a chymase inhibitor.
4078. The method of item 4055 wherein the agent is a collagenase antagonist.
4079. The method of item 4055 wherein the agent is a CXCR antagonist.
4080. The method of item 4055 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4081. The method of item 4055 wherein the agent is a cyclooxygenase 1 inhibitor.
4082. The method of item 4055 wherein the agent is a DHFR inhibitor.
4083. The method of item 4055 wherein the agent is a dual integrin inhibitor.
4084. The method of item 4055 wherein the agent is an elastase inhibitor.
4085. The method of item 4055 wherein the agent is an elongation factor-1 alpha inhibitor. 4086. The method of item 4055 wherein the agent is an endothelial growth factor antagonist.
4087. The method of item 4055 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4088. The method of item 4055 wherein the agent is an endotoxin antagonist.
4089. The method of item 4055 wherein the agent is an epothilone and tubulin binder.
4090. The method of item 4055 wherein the agent is an estrogen receptor antagonist.
4091. The method of item 4055 wherein the agent is an FGF inhibitor.
4092. The method of item 4055 wherein the agent is a farnexyl transferase inhibitor.
4093. The method of item 4055 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4094. The method of item 4055 wherein the agent is an FLT-3 kinase inhibitor.
4095. The method of item 4055 wherein the agent is an FGF receptor kinase inhibitor.
4096. The method of item 4055 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4097. The method of item 4055 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
4098. The method of item 4055 wherein the agent is a histone deacetylase inhibitor.
4099. The method of item 4055 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 4100. The method of item 4055 wherein the agent is an ICAM inhibitor.
4101. The method of item 4055 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
4102. The method of item 4055 wherein the agent is an IL-2 inhibitor.
4103. The method of item 4055 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4104. The method of item 4055 wherein the agent is an IMPDH (inosine monophosphate). 4105. The method of item 4055 wherein the agent is an integrin antagonist.
4106. The method of item 4055 wherein the agent is an interleukin antagonist.
4107. The method of item 4055 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
4108. The method of item 4055 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4109. The method of item 4055 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
4110. The method of item 4055 wherein the agent a JAK3 enzyme inhibitor.
4111. The method of item 4055 wherein the agent is a JNK inhibitor.
4112. The method of item 4055 wherein the agent is a kinase inhibitor.
4113. The method of item 4055 wherein the agent is kinesin antagonist.
4114. The method of item 4055 wherein the agent is a kinesin antagonist.
4115. The method of item 4055 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW- 1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No.D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4116. The method of item 4055 wherein the agent is an MAP kinase inhibitor.
4117. The method of item 4055 wherein the agent is a matrix metalloproteinase inhibitor.
4118. The method of item 4055 wherein the agent is an MCP-
CCR2 inhibitor.
4119. The method of item 4055 wherein the agent is an mTOR inhibitor.
4120. The method of item 4055 wherein the agent is an mTOR kinase inhibitor.
4121. The method of item 4055 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
4122. The method of item 4055 wherein the agent is an MIF inhibitor.
4123. The method of item 4055 wherein the agent is an MMP inhibitor.
4124. The method of item 4055 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4125. The method of item 4055 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 4126. The method of item 4055 wherein the agent is a nitric oxide agonist.
4127. The method of item 4055 wherein the agent is an ornithine decarboxylase inhibitor.
4128. The method of item 4055 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4129. The method of item 4055 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4130. The method of item 4055 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4131. The method of item 4055 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + piogiitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, piogiitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4132. The method of item 4055 wherein the agent is a phosphatase inhibitor.
4133. The method of item 4055 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV
» inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (in2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
4134. The method of item 4055 wherein the agent is a PKC inhibitor.
4135. The method of item 4055 wherein the agent is a platelet activating factor antagonist.
4136. The method of item 4055 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4137. The method of item 4055 wherein the agent is a prolyl hydroxylase inhibitor.
4138. The method of item 4055 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4139. The method of item 4055 wherein the agent is a protein kinase B inhibitor.
4140. The method of item 4055 wherein the agent is a protein kinase C stimulant.
4141. The method of item 4055 wherein the agent is a purine nucleoside analogue.
4142. The method of item 4055 wherein the agent is a purinoreceptor P2X antagonist.
4143. The method of item 4055 wherein the agent is a Raf kinase inhibitor. 4144. The method of item 4055 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4145. The method of item 4055 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
4146. The method of item 4055 wherein the agent is an SDF-1 antagonist.
4147. The method of item 4055 wherein the agent is a sheddase inhibitor.
4148. The method of item 4055 wherein the agent is an SRC inhibitor.
4149. The method of item 4055 wherein the agent is a stromelysin inhibitor.
4150. The method of item 4055 wherein the agent is an Syk kinase inhibitor.
4151. The method of item 4055 wherein the agent is a telomerase inhibitor.
4152. The method of item 4055 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4153. The method of item 4055 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, antiinflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB)1 dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y1S Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
4154. The method of item 4055 wherein the agent is a Toll receptor inhibitor.
4155. The method of item 4055 wherein the agent is a tubulin antagonist.
4156. The method of item 4055 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4157. The method of item 4055 wherein the agent is a VEGF inhibitor.
4158. The method of item 4055 wherein the agent is a vitamin D receptor agonist.
4159. The method of item 4055 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
4160. The method of item 4055 wherein the agent is AP-23573 (an mTOR inhibitor). 4161. The method of item 4055 wherein the agent is synthadotin (a tubulin antagonist).
4162. The method of item 4055 wherein the agent is S-0885 (a collagenase inhibitor).
4163. The method of item 4055 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
4164. The method of item 4055 wherein the agent is ixabepilone (an epithilone).
4165. The method of item 4055 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
4166. The method of item 4055 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4167. The method of item 4055 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4168. The method of item 4055 wherein the agent is combretastatin (an angiogenesis inhibitor).
4169. The method of item 4055 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4170. The method of item 4055 wherein the agent is SB- 715992 (a kinesin antagonist).
4171. The method of item 4055 wherein the agent is temsirolimus (an mTOR inhibitor).
4172. The method of item 4055 wherein the agent is adalimumab (a TNFα antagonist). 4173. The method of item 4055, wherein the composition comprises a polymer.
4174. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
4175. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
4176. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
4177. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
4178. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
4179. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
4180. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
4181. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
4182. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 4183. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
4184. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
4185. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
4186. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
4187. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
4188. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
4189. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
4190. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
4191. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
4192. The method of item 4055, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
4193. The method of item 4055, wherein the composition further comprises a second pharmaceutically active agent. 4194. The method of item 4055, wherein the composition further comprises an anti-inflammatory agent.
4195. The method of item 4055, wherein the composition further comprises an agent that inhibits infection.
4196. The method of item 4055, wherein the composition further comprises an anthracycline.
4197. The method of item 4055, wherein the composition further comprises doxorubicin.
4198. The method of item 4055 wherein the composition further comprises mitoxantrone.
4199. The method of item 4055 wherein the composition further comprises a fluoropyrimidine.
4200. The method of item 4055, wherein the composition further comprises 5-fluorouracil (5-FU).
4201. The method of item 4055, wherein the composition further comprises a folic acid antagonist.
4202. The method of item 4055, wherein the composition further comprises methotrexate.
4203. The method of item 4055, wherein the composition further comprises a podophylotoxin.
4204. The method of item 4055, wherein the composition further comprises etoposide.
4205. The method of item 4055, wherein the composition further comprises camptothecin. 4206. The method of item 4055, wherein the composition further comprises a hydroxyurea.
4207. The method of item 4055, wherein the composition further comprises a platinum complex.
4208. The method of item 4055, wherein the composition further comprises cisplatin.
4209. The method of item 4055 wherein the composition further comprises an antithrombotic agent.
4210. The method of item 4055, wherein the composition further comprises a visualization agent.
4211. The method of item 4055, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4212. The method of item 4055, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
4213. The method of item 4055, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
4214. The method of item 4055, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
4215. The method of item 4055, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 4216. The method of item 4055, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
4217. The method of item 4055, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
4218. The method of item 4055 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
4219. The method of item 4055 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
4220. The method of item 4055 wherein the composition further comprises an inflammatory cytokine.
4221. The method of item 4055 wherein the composition further comprises an agent that stimulates cell proliferation.
4222. The method of item 4055 wherein the composition further comprises a polymeric carrier.
4223. The method of item 4055 wherein the composition is in the form of a gel, paste, or spray.
4224. The method of item 4055 wherein the sensor is partially constructed with the agent or the composition.
4225. The method of item 4055 wherein the sensor is impregnated with the agent or the composition. 4226. The method of item 4055, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
4227. The method of item 4055, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
4228. The method of item 4055 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
4229. The method of item 4055, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
4230. The method of item 4055 wherein the agent or the composition is located within pores or holes of the sensor.
4231. The method of item 4055 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
4232. The method of item 4055 wherein the sensor further comprises an echogenic material.
4233. The method of item 4055 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
4234. The method of item 4055 wherein the sensor is sterile.
4235. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
4236. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue. 4237. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
4238. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
4239. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
4240. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
4241. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
4242. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
4243. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
4244. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate. 4245. The method of item 4055 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
4246. The method of item 4055 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
4247. The method of item 4055 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
4248. The method of item 4055 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
4249. The method of item 4055 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
4250. The method of item 4055 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
4251. The method of item 4055 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
4252. The method of item 4055 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied. 4253. The method of item 4055 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
4254. The method of item 4055 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
4255. The method of item 4055 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
4256. The method of item 4055 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
4257. The method of item 4055, wherein the sensor further comprises a coating, and the coating is a uniform coating.
4258. The method of item 4055, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
4259. The method of item 4055, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
4260. The method of item 4055, wherein the sensor further comprises a coating, and the coating is a patterned coating.
4261. The method of item 4055, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less. 4262. The method of item 4055, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
4263. The method of item 4055, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
4264. The method of item 4055, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
4265. The method of item 4055, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4266. The method of item 4055, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4267. The method of item 4055, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4268. The method of item 4055, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4269. The method of item 4055, wherein the sensor further comprises a coating, and the coating comprises a polymer.
4270. The method of item 4055, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition. 4271. The method of item 4055, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
4272. The method of item 4055 wherein the agent or the composition is affixed to the sensor.
4273. The method of item 4055 wherein the agent or the composition is covalently attached to the sensor.
4274. The method of item 4055 wherein the agent or the composition is non-covalently attached to the sensor.
4275. The method of item 4055 wherein the sensor comprises a coating that absorbs the agent or the composition.
4276. The method of item 4055 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
4277. The method of item 4055 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
4278. The method of item 4055 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
4279. The method of item 4055 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
4280. The method of item 4055 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
4281. The method of item 4055 wherein the sensor is linked to a pump. 4282. The method of item 4055 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
4283. The method of item 4055 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
4284. The method of item 4055 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
4285. The method of item 4055 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
4286. The method of item 4055 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
4287. The method of item 4055 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
4288. The method of item 4055 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
4289. The method of item 4055 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
4290. The method of any one of items 4055-4289 wherein the sensor monitors pulmonary functions. 4291. A method for inhibiting scarring comprising placing an auditory sensor and an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
4292. The method of item 4291 wherein the agent is an adensosine A2A receptor antagonist.
4293. The method of item 4291 wherein the agent is an AKT inhibitor.
4294. The method of item 4291 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4295. The method of item 4291 wherein the agent is an alpha 4 integrin antagonist.
4296. The method of item 4291 wherein the agent is an alpha 7 nicotinic receptor agonist.
4297. The method of item 4291 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma),' Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4298. The method of item 4291 wherein the agent is an apoptosis antagonist.
4299. The method of item 4291 wherein the agent is an apoptosis activator.
4300. The method of item 4291 wherein the agent is a beta 1 integrin antagonist.
4301. The method of item 4291 wherein the agent is a beta tubulin inhibitor.
4302. The method of item 4291 wherein the agent is a blocker of enzyme production in Hepatitis C.
4303. The method of item 4291 wherein the agent is a Bruton's tyrosine kinase inhibitor. 4304. The method of item 4291 wherein the agent is a calcineurin inhibitor.
4305. The method of item 4291 wherein the agent is a caspase 3 inhibitor.
4306. The method of item 4291 wherein the agent is a CC chemokine receptor antagonist.
4307. The method of item 4291 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4308. The method of item 4291 wherein the agent is a cathepsin B inhibitor.
4309. The method of item 4291 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
4310. The method of item 4291 wherein the agent is a cathepsin L inhibitor.
4311. The method of item 4291 wherein the agent is a CD40 antagonist.
4312. The method of item 4291 wherein the agent is a chemokine receptor agonist.
4313. The method of item 4291 wherein the agent is a chymase inhibitor.
4314. The method of item 4291 wherein the agent is a collagenase antagonist. 4315. The method of item 4291 wherein the agent is a CXCR antagonist.
4316. The method of item 4291 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4317. The method of item 4291 wherein the agent is a cyclooxygenase 1 inhibitor.
4318. The method of item 4291 wherein the agent is a DHFR inhibitor.
4319. The method of item 4291 wherein the agent is a dual integrin inhibitor.
4320. The method of item 4291 wherein the agent is an elastase inhibitor.
4321. The method of item 4291 wherein the agent is an elongation factor-1 alpha inhibitor.
4322. The method of item 4291 wherein the agent is an endothelial growth factor antagonist.
4323. The method of item 4291 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4324. The method of item 4291 wherein the agent is an endotoxin antagonist.
4325. The method of item 4291 wherein the agent is an epothilone and tubulin binder.
4326. The method of item 4291 wherein the agent is an estrogen receptor antagonist.
4327. The method of item 4291 wherein the agent is an FGF inhibitor.
4328. The method of item 4291 wherein the agent is a famexyl transferase inhibitor.
4329. The method of item 4291 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4330. The method of item 4291 wherein the agent is an FLT-3 kinase inhibitor. 4331. The method of item 4291 wherein the agent is an FGF receptor kinase inhibitor.
4332. The method of item 4291 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4333. The method of item 4291 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005
(Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
4334. The method of item 4291 wherein the agent is a histone deacetylase inhibitor.
4335. The method of item 4291 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
4336. The method of item 4291 wherein the agent is an ICAM inhibitor.
4337. The method of item 4291 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 4338. The method of item 4291 wherein the agent is an IL-2 inhibitor.
4339. The method of item 4291 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4340. The method of item 4291 wherein the agent is an IMPDH (inosine monophosphate).
4341. The method of item 4291 wherein the agent is an integrin antagonist.
4342. The method of item 4291 wherein the agent is an interleukin antagonist.
4343. The method of item 4291 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 4344. The method of item 4291 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4345. The method of item 4291 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
4346. The method of item 4291 wherein the agent a JAK3 enzyme inhibitor.
4347. The method of item 4291 wherein the agent is a JNK inhibitor.
4348. The method of item 4291 wherein the agent is a kinase inhibitor.
4349. The method of item 4291 wherein the agent is kinesin antagonist.
4350. The method of item 4291 wherein the agent is a kinesin antagonist.
4351. The method of item 4291 wherein the agent is a leυkotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4352. The method of item 4291 wherein the agent is an MAP kinase inhibitor.
4353. The method of item 4291 wherein the agent is a matrix metalloproteinase inhibitor.
4354. The method of item 4291 wherein the agent is an MCP- CCR2 inhibitor.
4355. The method of item 4291 wherein the agent is an mTOR inhibitor.
4356. The method of item 4291 wherein the agent is an mTOR kinase inhibitor.
4357. The method of item 4291 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
4358. The method of item 4291 wherein the agent is an MIF inhibitor. 4359. The method of item 4291 wherein the agent is an MMP inhibitor.
4360. The method of item 4291 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4361. The method of item 4291 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
4362. The method of item 4291 wherein the agent is a nitric oxide agonist.
4363. The method of item 4291 wherein the agent is an ornithine decarboxylase inhibitor.
4364. The method of item 4291 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4365. The method of item 4291 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4366. The method of item 4291 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-
860 (UCB)1 E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4367. The method of item 4291 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET
(GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4368. The method of item 4291 wherein the agent is a phosphatase inhibitor.
4369. The method of item 4291 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 4370. The method of item 4291 wherein the agent is a PKC inhibitor.
4371. The method of item 4291 wherein the agent is a platelet activating factor antagonist.
4372. The method of item 4291 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4373. The method of item 4291 wherein the agent is a prolyl hydroxylase inhibitor.
4374. The method of item 4291 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4375. The method of item 4291 wherein the agent is a protein kinase B inhibitor.
4376. The method of item 4291 wherein the agent is a protein kinase C stimulant.
4377. The method of item 4291 wherein the agent is a purine nucleoside analogue.
4378. The method of item 4291 wherein the agent is a purinoreceptor P2X antagonist.
4379. The method of item 4291 wherein the agent is a Raf kinase inhibitor.
4380. The method of item 4291 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4381. The method of item 4291 wherein the agent is a ribonucleoside triphosphate reductase inhibitor. 4382. The method of item 4291 wherein the agent is an SDF-1 antagonist.
4383. The method of item 4291 wherein the agent is a sheddase inhibitor.
4384. The method of item 4291 wherein the agent is an SRC inhibitor.
4385. The method of item 4291 wherein the agent is a stromelysin inhibitor.
4386. The method of item 4291 wherein the agent is an Syk kinase inhibitor.
4387. The method of item 4291 wherein the agent is a telomerase inhibitor.
4388. The method of item 4291 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4389. The method of item 4291 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB)1 cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 4390. The method of item 4291 wherein the agent is a Toll receptor inhibitor.
4391. The method of item 4291 wherein the agent is a tubulin antagonist.
4392. The method of item 4291 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis
Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4393. The method of item 4291 wherein the agent is a VEGF inhibitor.
4394. The method of item 4291 wherein the agent is a vitamin D receptor agonist.
4395. The method of item 4291 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
4396. The method of item 4291 wherein the agent is AP-23573 (an mTOR inhibitor).
4397. The method of item 4291 wherein the agent is synthadotin (a tubulin antagonist).
4398. The method of item 4291 wherein the agent is S-0885 (a collagenase inhibitor). 4399. The method of item 4291 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
4400. The method of item 4291 wherein the agent is ixabepilone (an epithilone).
4401. The method of item 4291 wherein the agent is IDN-5390
(an angiogenesis inhibitor).
4402. The method of item 4291 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4403. The method of item 4291 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4404. The method of item 4291 wherein the agent is combretastatin (an angiogenesis inhibitor).
4405. The method of item 4291 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4406. The method of item 4291 wherein the agent is SB-
715992 (a kinesin antagonist).
4407. The method of item 4291 wherein the agent is temsirolimus (an mTOR inhibitor).
4408. The method of item 4291 wherein the agent is adalimumab (a TNFα antagonist).
4409. The method of item 4291 , wherein the composition comprises a polymer.
4410. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 4411. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
4412. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
4413. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
4414. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
4415. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
4416. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
4417. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
4418. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
4419. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
4420. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer. 4421. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
4422. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
4423. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
4424. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
4425. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
4426. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
4427. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
4428. The method of item 4291 , wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
4429. The method of item 4291 , wherein the composition further comprises a second pharmaceutically active agent.
4430. The method of item 4291 , wherein the composition further comprises an anti-inflammatory agent.
4431. The method of item 4291 , wherein the composition further comprises an agent that inhibits infection. 4432. The method of item 4291 , wherein the composition further comprises an anthracycline.
4433. The method of item 4291 , wherein the composition further comprises doxorubicin.
4434. The method of item 4291 wherein the composition further comprises mitoxantrone.
4435. The method of item 4291 wherein the composition further comprises a fluoropyrimidine.
4436. The method of item 4291 , wherein the composition further comprises 5-fluorouracil (5-FU).
4437. The method of item 4291 , wherein the composition further comprises a folic acid antagonist.
4438. The method of item 4291 , wherein the composition further comprises methotrexate.
4439. The method of item 4291 , wherein the composition further comprises a podophylotoxin.
4440. The method of item 4291 , wherein the composition further comprises etoposide.
4441. The method of item 4291 , wherein the composition further comprises camptothecin.
4442. The method of item 4291 , wherein the composition further comprises a hydroxyurea.
4443. The method of item 4291 , wherein the composition further comprises a platinum complex. 4444. The method of item 4291 , wherein the composition further comprises cisplatin.
4445. The method of item 4291 wherein the composition further comprises an anti-thrombotic agent.
4446. The method of item 4291 , wherein the composition further comprises a visualization agent.
4447. The method of item 4291 , wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4448. The method of item 4291 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
4449. The method of item 4291 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
4450. The method of item 4291 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
4451. The method of item 4291 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
4452. The method of item 4291 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound. 4453. The method of item 4291 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
4454. The method of item 4291 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
4455. The method of item 4291 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
4456. The method of item 4291 wherein the composition further comprises an inflammatory cytokine.
4457. The method of item 4291 wherein the composition further comprises an agent that stimulates cell proliferation.
4458. The method of item 4291 wherein the composition further comprises a polymeric carrier.
4459. The method of item 4291 wherein the composition is in the form of a gel, paste, or spray.
4460. The method of item 4291 wherein the sensor is partially constructed with the agent or the composition.
4461. The method of item 4291 wherein the sensor is impregnated with the agent or the composition.
4462. The method of item 4291 , wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
4463. The method of item 4291 , wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor. 4464. The method of item 4291 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
4465. The method of item 4291 , wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
4466. The method of item 4291 wherein the agent or the composition is located within pores or holes of the sensor.
4467. The method of item 4291 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
4468. The method of item 4291 wherein the sensor further comprises an echogenic material.
4469. The method of item 4291 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
4470. The method of item 4291 wherein the sensor is sterile.
4471. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
4472. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
4473. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
4474. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue. 4475. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
4476. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
4477. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
4478. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
4479. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
4480. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
4481. The method of item 4291 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
4482. The method of item 4291 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent. 4483. The method of item 4291 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
4484. The method of item 4291 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
4485. The method of item 4291 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
4486. The method of item 4291 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
4487. The method of item 4291 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
4488. The method of item 4291 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
4489. The method of item 4291 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
4490. The method of item 4291 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied. 4491. The method of item 4291 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
4492. The method of item 4291 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
4493. The method of item 4291 , wherein the sensor further comprises a coating, and the coating is a uniform coating.
4494. The method of item 4291 , wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
4495. The method of item 4291 , wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
4496. The method of item 4291 , wherein the sensor further comprises a coating, and the coating is a patterned coating.
4497. The method of item 4291 , wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
4498. The method of item 4291 , wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
4499. The method of item 4291 , wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
4500. The method of item 4291 , wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year. 4501. The method of item 4291 , wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4502. The method of item 4291 , wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4503. The method of item 4291 , wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4504. The method of item 4291 , wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4505. The method of item 4291 , wherein the sensor further comprises a coating, and the coating comprises a polymer.
4506. The method of item 4291 , wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
4507. The method of item 4291 , wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
4508. The method of item 4291 wherein the agent or the composition is affixed to the sensor.
4509. The method of item 4291 wherein the agent or the composition is covalently attached to the sensor. 4510. The method of item 4291 wherein the agent or the composition is non-covalently attached to the sensor.
4511. The method of item 4291 wherein the sensor comprises a coating that absorbs the agent or the composition.
4512. The method of item 4291 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
4513. The method of item 4291 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
4514. The method of item 4291 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
4515. The method of item 4291 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
4516. The method of item 4291 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
4517. The method of item 4291 wherein the sensor is linked to a pump.
4518. The method of item 4291 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
4519. The method of item 4291 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
4520. The method of item 4291 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host. 4521. The method of item 4291 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
4522. The method of item 4291 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
4523. The method of item 4291 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
4524. The method of item 4291 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
4525. The method of item 4291 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
4526. The method of any one of items 4291-4525 wherein the sensor is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
4527. The method of any one of items 4291-4525 wherein the sensor generates an electrical audio signal.
4528. The method of any one of items 4291-4525 wherein the sensor is a capacitive sensor that is coupled to a vibrating auditory element.
4529. The method of any one of items 4291-4525 wherein the sensor is an electromagnetic sensor.
4530. A method for inhibiting scarring comprising placing an electrolyte or metabolite sensor and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
4531. The method of item 4530 wherein the agent is an adensosine A2A receptor antagonist.
4532. The method of item 4530 wherein the agent is an AKT inhibitor.
4533. The method of item 4530 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4534. The method of item 4530 wherein the agent is an alpha 4 integrin antagonist.
4535. The method of item 4530 wherein the agent is an alpha 7 nicotinic receptor agonist.
4536. The method of item 4530 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW- 114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4537. The method of item 4530 wherein the agent is an apoptosis antagonist.
4538. The method of item 4530 wherein the agent is an apoptosis activator.
4539. The method of item 4530 wherein the agent is a beta 1 integrin antagonist.
4540. The method of item 4530 wherein the agent is a beta tubulin inhibitor.
4541. The method of item 4530 wherein the agent is a blocker of enzyme production in Hepatitis C.
4542. The method of item 4530 wherein the agent is a Bruton's tyrosine kinase inhibitor. 4543. The method of item 4530 wherein the agent is a calcineurin inhibitor.
4544. The method of item 4530 wherein the agent is a caspase 3 inhibitor.
4545. The method of item 4530 wherein the agent is a CC chemokine receptor antagonist.
4546. The method of item 4530 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4547. The method of item 4530 wherein the agent is a cathepsin B inhibitor.
4548. The method of item 4530 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
4549. The method of item 4530 wherein the agent is a cathepsin L inhibitor.
4550. The method of item 4530 wherein the agent is a CD40 antagonist.
4551. The method of item 4530 wherein the agent is a chemokine receptor agonist.
4552. The method of item 4530 wherein the agent is a chymase inhibitor.
4553. The method of item 4530 wherein the agent is a collagenase antagonist. 4554. The method of item 4530 wherein the agent is a CXCR antagonist.
4555. The method of item 4530 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4556. The method of item 4530 wherein the agent is a cyclooxygenase 1 inhibitor.
4557. The method of item 4530 wherein the agent is a DHFR inhibitor.
4558. The method of item 4530 wherein the agent is a dual integrin inhibitor.
4559. The method of item 4530 wherein the agent is an elastase inhibitor.
4560. The method of item 4530 wherein the agent is an elongation factor-1 alpha inhibitor.
4561. The method of item 4530 wherein the agent is an endothelial growth factor antagonist.
4562. The method of item 4530 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI
Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4563. The method of item 4530 wherein the agent is an endotoxin antagonist.
4564. The method of item 4530 wherein the agent is an epothilone and tubulin binder.
4565. The method of item 4530 wherein the agent is an estrogen receptor antagonist.
4566. The method of item 4530 wherein the agent is an FGF inhibitor.
4567. The method of item 4530 wherein the agent is a famexyl transferase inhibitor.
4568. The method of item 4530 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4569. The method of item 4530 wherein the agent is an FLT-3 kinase inhibitor. 4570. The method of item 4530 wherein the agent is an FGF receptor kinase inhibitor.
4571. The method of item 4530 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4572. The method of item 4530 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005
(Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1?,4-didehydro-1-deoxy-1 ,4~dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
4573. The method of item 4530 wherein the agent is a histone deacetylase inhibitor.
4574. The method of item 4530 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
4575. The method of item 4530 wherein the agent is an ICAM inhibitor.
4576. The method of item 4530 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 4577. The method of item 4530 wherein the agent is an IL-2 inhibitor.
4578. The method of item 4530 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4579. The method of item 4530 wherein the agent is an IMPDH (inosine monophosphate).
4580. The method of item 4530 wherein the agent is an integrin antagonist.
4581. The method of item 4530 wherein the agent is an interleukin antagonist.
4582. The method of item 4530 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 4583. The method of item 4530 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4584. The method of item 4530 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
4585. The method of item 4530 wherein the agent a JAK3 enzyme inhibitor.
4586. The method of item 4530 wherein the agent is a JNK inhibitor.
4587. The method of item 4530 wherein the agent is a kinase inhibitor.
4588. The method of item 4530 wherein the agent is kinesin antagonist.
4589. The method of item 4530 wherein the agent is a kinesin antagonist.
4590. The method of item 4530 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4591. The method of item 4530 wherein the agent is an MAP kinase inhibitor.
4592. The method of item 4530 wherein the agent is a matrix metalloproteinase inhibitor.
4593. The method of item 4530 wherein the agent is an MCP- CCR2 inhibitor.
4594. The method of item 4530 wherein the agent is an mTOR inhibitor.
4595. The method of item 4530 wherein the agent is an mTOR kinase inhibitor.
4596. The method of item 4530 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
4597. The method of item 4530 wherein the agent is an MIF inhibitor. 4598. The method of item 4530 wherein the agent is an MMP inhibitor.
4599. The method of item 4530 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4600. The method of item 4530 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
4601. The method of item 4530 wherein the agent is a nitric oxide agonist.
4602. The method of item 4530 wherein the agent is an ornithine decarboxylase inhibitor.
4603. The method of item 4530 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4604. The method of item 4530 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4605. The method of item 4530 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4606. The method of item 4530 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET
(GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4607. The method of item 4530 wherein the agent is a phosphatase inhibitor.
4608. The method of item 4530 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimiiast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 4609. The method of item 4530 wherein the agent is a PKC inhibitor.
4610. The method of item 4530 wherein the agent is a platelet activating factor antagonist.
4611. The method of item 4530 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4612. The method of item 4530 wherein the agent is a prolyl hydroxylase inhibitor.
4613. The method of item 4530 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4614. The method of item 4530 wherein the agent is a protein kinase B inhibitor.
4615. The method of item 4530 wherein the agent is a protein kinase C stimulant.
4616. The method of item 4530 wherein the agent is a purine nucleoside analogue.
4617. The method of item 4530 wherein the agent is a purinoreceptor P2X antagonist.
4618. The method of item 4530 wherein the agent is a Raf kinase inhibitor.
4619. The method of item 4530 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4620. The method of item 4530 wherein the agent is a ribonucleoside triphosphate reductase inhibitor. 4621. The method of item 4530 wherein the agent is an SDF-1 antagonist.
4622. The method of item 4530 wherein the agent is a sheddase inhibitor.
4623. The method of item 4530 wherein the agent is an SRC inhibitor.
4624. The method of item 4530 wherein the agent is a stromelysin inhibitor.
4625. The method of item 4530 wherein the agent is an Syk kinase inhibitor.
4626. The method of item 4530 wherein the agent is a telomerase inhibitor.
4627. The method of item 4530 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4628. The method of item 4530 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGlX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 4629. The method of item 4530 wherein the agent is a Toll receptor inhibitor.
4630. The method of item 4530 wherein the agent is a tubulin antagonist.
4631. The method of item 4530 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis
Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4632. The method of item 4530 wherein the agent is a VEGF inhibitor.
4633. The method of item 4530 wherein the agent is a vitamin D receptor agonist.
4634. The method of item 4530 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
4635. The method of item 4530 wherein the agent is AP-23573 (an mTOR inhibitor).
4636. The method of item 4530 wherein the agent is synthadotin (a tubulin antagonist).
4637. The method of item 4530 wherein the agent is S-0885 (a collagenase inhibitor). 4638. The method of item 4530 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
4639. The method of item 4530 wherein the agent is ixabepilone (an epithilone).
4640. The method of item 4530 wherein the agent is IDN-5390
(an angiogenesis inhibitor).
4641. The method of item 4530 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4642. The method of item 4530 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4643. The method of item 4530 wherein the agent is combretastatin (an angiogenesis inhibitor).
4644. The method of item 4530 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4645. The method of item 4530 wherein the agent is SB-
715992 (a kinesin antagonist).
4646. The method of item 4530 wherein the agent is temsirolimus (an mTOR inhibitor).
4647. The method of item 4530 wherein the agent is adalimumab (a TNFα antagonist).
4648. The method of item 4530, wherein the composition comprises a polymer.
4649. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 4650. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
4651. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
4652. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
4653. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
4654. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
4655. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
4656. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
4657. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
4658. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
4659. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer. 4660. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
4661. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
4662. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
4663. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
4664. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
4665. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
4666. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
4667. The method of item 4530, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
4668. The method of item 4530, wherein the composition further comprises a second pharmaceutically active agent.
4669. The method of item 4530, wherein the composition further comprises an anti-inflammatory agent.
4670. The method of item 4530, wherein the composition further comprises an agent that inhibits infection. 4671. The method of item 4530, wherein the composition further comprises an anthracycline.
4672. The method of item 4530, wherein the composition further comprises doxorubicin.
4673. The method of item 4530 wherein the composition further comprises mitoxantrone.
4674. The method of item 4530 wherein the composition further comprises a fluoropyrimidine.
4675. The method of item 4530, wherein the composition further comprises 5-fluorouracil (5-FU).
4676. The method of item 4530, wherein the composition further comprises a folic acid antagonist.
4677. The method of item 4530, wherein the composition further comprises methotrexate.
4678. The method of item 4530, wherein the composition further comprises a podophylotoxin.
4679. The method of item 4530, wherein the composition further comprises etoposide.
4680. The method of item 4530, wherein the composition further comprises camptothecin.
4681. The method of item 4530, wherein the composition further comprises a hydroxyurea.
4682. The method of item 4530, wherein the composition further comprises a platinum complex. 4683. The method of item 4530, wherein the composition further comprises cisplatin.
4684. The method of item 4530 wherein the composition further comprises an anti-thrombotic agent.
4685. The method of item 4530, wherein the composition further comprises a visualization agent.
4686. The method of item 4530, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4687. The method of item 4530, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
4688. The method of item 4530, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
4689. The method of item 4530, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
4690. The method of item 4530, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
4691. The method of item 4530, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound. 4692. The method of item 4530, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
4693. The method of item 4530 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
4694. The method of item 4530 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
4695. The method of item 4530 wherein the composition further comprises an inflammatory cytokine.
4696. The method of item 4530 wherein the composition further comprises an agent that stimulates cell proliferation.
4697. The method of item 4530 wherein the composition further comprises a polymeric carrier.
4698. The method of item 4530 wherein the composition is in the form of a gel, paste, or spray.
4699. The method of item 4530 wherein the sensor is partially constructed with the agent or the composition.
4700. The method of item 4530 wherein the sensor is impregnated with the agent or the composition.
4701. The method of item 4530, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
4702. The method of item 4530, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor. 4703. The method of item 4530 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
4704. The method of item 4530, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
4705. The method of item 4530 wherein the agent or the composition is located within pores or holes of the sensor.
4706. The method of item 4530 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
4707. The method of item 4530 wherein the sensor further comprises an echogenic material.
4708. The method of item 4530 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
4709. The method of item 4530 wherein the sensor is sterile.
4710. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
4711. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
4712. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
4713. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue. 4714. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
4715. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
4716. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
4717. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
4718. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
4719. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
4720. The method of item 4530 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
4721. The method of item 4530 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent. 4722. The method of item 4530 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
4723. The method of item 4530 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
4724. The method of item 4530 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
4725. The method of item 4530 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
4726. The method of item 4530 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
4727. The method of item 4530 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
4728. The method of item 4530 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
4729. The method of item 4530 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied. 4730. The method of item 4530 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
4731. The method of item 4530 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
4732. The method of item 4530, wherein the sensor further comprises a coating, and the coating is a uniform coating.
4733. The method of item 4530, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
4734. The method of item 4530, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
4735. The method of item 4530, wherein the sensor further comprises a coating, and the coating is a patterned coating.
4736. The method of item 4530, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
4737. The method of item 4530, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
4738. The method of item 4530, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
4739. The method of item 4530, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year. 4740. The method of item 4530, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4741. The method of item 4530, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4742. The method of item 4530, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4743. The method of item 4530, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4744. The method of item 4530, wherein the sensor further comprises a coating, and the coating comprises a polymer.
4745. The method of item 4530, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
4746. The method of item 4530, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
4747. The method of item 4530 wherein the agent or the composition is affixed to the sensor.
4748. The method of item 4530 wherein the agent or the composition is covalently attached to the sensor. 4749. The method of item 4530 wherein the agent or the composition is non-covalently attached to the sensor.
4750. The method of item 4530 wherein the sensor comprises a coating that absorbs the agent or the composition.
4751. The method of item 4530 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
4752. The method of item 4530 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
4753. The method of item 4530 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
4754. The method of item 4530 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
4755. The method of item 4530 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
4756. The method of item 4530 wherein the sensor is linked to a pump.
4757. The method of item 4530 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
4758. The method of item 4530 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
4759. The method of item 4530 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host. 4760. The method of item 4530 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
4761. The method of item 4530 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
4762. The method of item 4530 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
4763. The method of item 4530 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
4764. The method of item 4530 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
4765. The method of any one of items 4530-4764 wherein the sensor emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
4766. The method of any one of items 4530-4764 wherein the sensor is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader.
4767. The method of any one of items 4530-4764 wherein the sensor is a monolithic bioelectronic device for detecting at least one analyte within the host. 4768. A method for inhibiting scarring comprising placing a pump and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
4769. The method of item' 4768 wherein the agent is an adensosine A2A receptor antagonist.
4770. The method of item 4768 wherein the agent is an AKT inhibitor.
4771. The method of item 4768 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4772. The method of item 4768 wherein the agent is an alpha 4 integrin antagonist.
4773. The method of item 4768 wherein the agent is an alpha 7 nicotinic receptor agonist.
4774. The method of item 4768 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Sa!medix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wiiex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4775. The method of item 4768 wherein the agent is an apoptosis antagonist.
4776. The method of item 4768 wherein the agent is an apoptosis activator.
4777. The method of item 4768 wherein the agent is a beta 1 integrin antagonist.
4778. The method of item 4768 wherein the agent is a beta tubulin inhibitor.
4779. The method of item 4768 wherein the agent is a blocker of enzyme production in Hepatitis C.
4780. The method of item 4768 wherein the agent is a Bruton's tyrosine kinase inhibitor. 4781. The method of item 4768 wherein the agent is a calcineurin inhibitor.
4782. The method of item 4768 wherein the agent is a caspase 3 inhibitor.
4783. The method of item 4768 wherein the agent is a CC chemokine receptor antagonist.
4784. The method of item 4768 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4785. The method of item 4768 wherein the agent is a cathepsin B inhibitor.
4786. The method of item 4768 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
4787. The method of item 4768 wherein the agent is a cathepsin L inhibitor.
4788. The method of item 4768 wherein the agent is a CD40 antagonist.
4789. The method of item 4768 wherein the agent is a chemokine receptor agonist.
4790. The method of item 4768 wherein the agent is a chymase inhibitor.
4791. The method of item 4768 wherein the agent is a collagenase antagonist. 4792. The method of item 4768 wherein the agent is a CXCR antagonist.
4793. The method of item 4768 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4794. The method of item 4768 wherein the agent is a cyclooxygenase 1 inhibitor.
4795. The method of item 4768 wherein the agent is a DHFR inhibitor.
4796. The method of item 4768 wherein the agent is a dual integrin inhibitor.
4797. The method of item 4768 wherein the agent is an elastase inhibitor.
4798. The method of item 4768 wherein the agent is an elongation factor-1 alpha inhibitor.
4799. The method of item 4768 wherein the agent is an endothelial growth factor antagonist.
4800. The method of item 4768 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI
Pharmaceuticals), SP-5.2 (Supratek Pharma), SLM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4801. The method of item 4768 wherein the agent is an endotoxin antagonist.
4802. The method of item 4768 wherein the agent is an epothilone and tubulin binder.
4803. The method of item 4768 wherein the agent is an estrogen receptor antagonist.
4804. The method of item 4768 wherein the agent is an FGF inhibitor.
4805. The method of item 4768 wherein the agent is a farnexyl transferase inhibitor.
4806. The method of item 4768 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4807. The method of item 4768 wherein the agent is an FLT-3 kinase inhibitor. 4808. The method of item 4768 wherein the agent is an FGF receptor kinase inhibitor.
4809. The method of item 4768 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4810. The method of item 4768 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005
(Sirenade), geldanamycin, NSC-33050 (17-aIlylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
4811. The method of item 4768 wherein the agent is a histone deacetylase inhibitor.
4812. The method of item 4768 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
4813. The method of item 4768 wherein the agent is an ICAM inhibitor.
4814. The method of item 4768 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 4815. The method of item 4768 wherein the agent is an IL-2 inhibitor.
4816. The method of item 4768 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4817. The method of item 4768 wherein the agent is an IMPDH (inosine monophosphate).
4818. The method of item 4768 wherein the agent is an integrin antagonist.
4819. The method of item 4768 wherein the agent is an interleukin antagonist.
4820. The method of item 4768 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 4821. The method of item 4768 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4822. The method of item 4768 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
4823. The method of item 4768 wherein the agent a JAK3 enzyme inhibitor.
4824. The method of item 4768 wherein the agent is a JNK inhibitor.
4825. The method of item 4768 wherein the agent is a kinase inhibitor.
4826. The method of item 4768 wherein the agent is kinesin antagonist.
4827. The method of item 4768 wherein the agent is a kinesin antagonist.
4828. The method of item 4768 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4829. The method of item 4768 wherein the agent is an MAP kinase inhibitor.
4830. The method of item 4768 wherein the agent is a matrix metalloproteinase inhibitor.
4831. The method of item 4768 wherein the agent is an MCP- CCR2 inhibitor.
4832. The method of item 4768 wherein the agent is an mTOR inhibitor.
4833. The method of item 4768 wherein the agent is an mTOR kinase inhibitor.
4834. The method of item 4768 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
4835. The method of item 4768 wherein the agent is an MIF inhibitor. 4836. The method of item 4768 wherein the agent is an MMP inhibitor.
4837. The method of item 4768 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1 ) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4838. The method of item 4768 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
4839. The method of item 4768 wherein the agent is a nitric oxide agonist.
4840. The method of item 4768 wherein the agent is an ornithine decarboxylase inhibitor.
4841. The method of item 4768 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4842. The method of item 4768 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4843. The method of item 4768 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-
860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4844. The method of item 4768 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET
(GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4845. The method of item 4768 wherein the agent is a phosphatase inhibitor.
4846. The method of item 4768 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 4847. The method of item 4768 wherein the agent is a PKC inhibitor.
4848. The method of item 4768 wherein the agent is a platelet activating factor antagonist.
4849. The method of item 4768 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4850. The method of item 4768 wherein the agent is a prolyl hydroxylase inhibitor.
4851. The method of item 4768 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4852. The method of item 4768 wherein the agent is a protein kinase B inhibitor.
4853. The method of item 4768 wherein the agent is a protein kinase C stimulant.
4854. The method of item 4768 wherein the agent is a purine nucleoside analogue.
4855. The method of item 4768 wherein the agent is a purinoreceptor P2X antagonist.
4856. The method of item 4768 wherein the agent is a Raf kinase inhibitor.
4857. The method of item 4768 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4858. The method of item 4768 wherein the agent is a ribonucleoside triphosphate reductase inhibitor. 4859. The method of item 4768 wherein the agent is an SDF-1 antagonist.
4860. The method of item 4768 wherein the agent is a sheddase inhibitor.
4861. The method of item 4768 wherein the agent is an SRC inhibitor.
4862. The method of item 4768 wherein the agent is a stromelysin inhibitor.
4863. The method of item 4768 wherein the agent is an Syk kinase inhibitor.
4864. The method of item 4768 wherein the agent is a telomerase inhibitor.
4865. The method of item 4768 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4866. The method of item 4768 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 4867. The method of item 4768 wherein the agent is a Toll receptor inhibitor.
4868. The method of item 4768 wherein the agent is a tubulin antagonist.
4869. The method of item 4768 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis
Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4870. The method of item 4768 wherein the agent is a VEGF inhibitor.
4871. The method of item 4768 wherein the agent is a vitamin D receptor agonist.
4872. The method of item 4768 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
4873. The method of item 4768 wherein the agent is AP-23573 (an mTOR inhibitor).
4874. The method of item 4768 wherein the agent is synthadotin (a tubulin antagonist).
4875. The method of item 4768 wherein the agent is S-0885 (a collagenase inhibitor). 4876. The method of item 4768 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
4877. The method of item 4768 wherein the agent is ixabepilone (an epithilone).
4878. The method of item 4768 wherein the agent is IDN-5390
(an angiogenesis inhibitor).
4879. The method of item 4768 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4880. The method of item 4768 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4881. The method of item 4768 wherein the agent is combretastatin (an angiogenesis inhibitor).
4882. The method of item 4768 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4883. The method of item 4768 wherein the agent is SB-
715992 (a kinesin antagonist).
4884. The method of item 4768 wherein the agent is temsirolimus (an mTOR inhibitor).
4885. The method of item 4768 wherein the agent is adalimumab (a TNFα antagonist).
4886. The method of item 4768, wherein the composition comprises a polymer.
4887. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 4888. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
4889. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
4890. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
4891. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
4892. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
4893. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
4894. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
4895. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
4896. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
4897. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer. 4898. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
4899. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
4900. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
4901. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
4902. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
4903. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
4904. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
4905. The method of item 4768, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
4906. The method of item 4768, wherein the composition further comprises a second pharmaceutically active agent.
4907. The method of item 4768, wherein the composition further comprises an anti-inflammatory agent.
4908. The method of item 4768, wherein the composition further comprises an agent that inhibits infection. 4909. The method of item 4768, wherein the composition further comprises an anthracycline.
4910. The method of item 4768, wherein the composition further comprises doxorubicin.
4911. The method of item 4768 wherein the composition further comprises mitoxantrone.
4912. The method of item 4768 wherein the composition further comprises a fluoropyrimidine.
4913. The method of item 4768, wherein the composition further comprises 5-fluorouracil (5-FU).
4914. The method of item 4768, wherein the composition further comprises a folic acid antagonist.
4915. The method of item 4768, wherein the composition further comprises methotrexate.
4916. The method of item 4768, wherein the composition further comprises a podophylotoxin.
4917. The method of item 4768, wherein the composition further comprises etoposide.
4918. The method of item 4768, wherein the composition further comprises camptothecin.
4919. The method of item 4768, wherein the composition further comprises a hydroxyurea.
4920. The method of item 4768, wherein the composition further comprises a platinum complex. 4921. The method of item 4768, wherein the composition further comprises cisplatin.
4922. The method of item 4768 wherein the composition further comprises an anti-thrombotic agent.
4923. The method of item 4768, wherein the composition further comprises a visualization agent.
4924. The method of item 4768, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4925. The method of item 4768, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
4926. The method of item 4768, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
4927. The method of item 4768, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
4928. The method of item 4768, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
4929. The method of item 4768, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound. 4930. The method of item 4768, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
4931. The method of item 4768 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
4932. The method of item 4768 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
4933. The method of item 4768 wherein the composition further comprises an inflammatory cytokine.
4934. The method of item 4768 wherein the composition further comprises an agent that stimulates cell proliferation.
4935. The method of item 4768 wherein the composition further comprises a polymeric carrier.
4936. The method of item 4768 wherein the composition is in the form of a gel, paste, or spray.
4937. The method of item 4768 wherein the sensor is partially constructed with the agent or the composition.
4938. The method of item 4768 wherein the sensor is impregnated with the agent or the composition.
4939. The method of item 4768, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
4940. The method of item 4768, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor. 4941. The method of item 4768 wherein the agent or the composition forms a coating, and the .coating partially covers the sensor.
4942. The method of item 4768, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
4943. The method of item 4768 wherein the agent or the composition is located within pores or holes of the sensor.
4944. The method of item 4768 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
4945. The method of item 4768 wherein the sensor further comprises an echogenic material.
4946. The method of item 4768 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
4947. The method of item 4768 wherein the sensor is sterile.
4948. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
4949. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
4950. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
4951. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue. 4952. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
4953. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
4954. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
4955. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
4956. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
4957. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
j 4958. The method of item 4768 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
4959. The method of item 4768 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent. 4960. The method of item 4768 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
4961. The method of item 4768 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
4962. The method of item 4768 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
4963. The method of item 4768 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
4964. The method of item 4768 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
4965. The method of item 4768 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
4966. The method of item 4768 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
4967. The method of item 4768 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied. 4968. The method of item 4768 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
4969. The method of item 4768 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
4970. The method of item 4768, wherein the sensor further comprises a coating, and the coating is a uniform coating.
4971. The method of item 4768, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
4972. The method of item 4768, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
4973. The method of item 4768, wherein the sensor further comprises a coating, and the coating is a patterned coating.
4974. The method of item 4768, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
4975. The method of item 4768, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
4976. The method of item 4768, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
4977. The method of item 4768, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year. 4978. The method of item 4768, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4979. The method of item 4768, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4980. The method of item 4768, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4981. The method of item 4768, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4982. The method of item 4768, wherein the sensor further comprises a coating, and the coating comprises a polymer.
4983. The method of item 4768, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
4984. The method of item 4768, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
4985. The method of item 4768 wherein the agent or the composition is affixed to the sensor.
4986. The method of item 4768 wherein the agent or the composition is covalently attached to the sensor. 4987. The method of item 4768 wherein the agent or the composition is non-covalently attached to the sensor.
4988. The method of item 4768 wherein the sensor comprises a coating that absorbs the agent or the composition.
4989. The method of item 4768 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
4990. The method of item 4768 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
4991. The method of item 4768 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
4992. The method of item 4768 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
4993. The method of item 4768 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
4994. The method of item 4768 wherein the sensor is linked to a pump.
4995. The method of item 4768 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
4996. The method of item 4768 wherein the agent or the composition is applied to the sensor surface during the placing, of the sensor into the host.
4997. The method of item 4768 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host. 4998. The method of item 4768 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
4999. The method of item 4768 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
5000. The method of item 4768 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
5001. The method of item 4768 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
5002. The method of item 4768 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
5003. The method of any one of items 4768-5002 wherein the pump is adapted for delivering insulin.
5004. The method of any one of items 4768-5002 wherein the pump is adapted for delivering a narcotic.
5005. The method of any one of items 4768-5002 wherein the pump is adapted for delivering a chemotherapeutic agent.
5006. The method of any one of items 4768-5002 wherein the pump is adapted for delivering an anti-arrhythmic drug.
5007. The method of any one of items 4768-5002 wherein the pump is adapted for delivering an anti-spasmotic drug. 5008. The method of any one of items 4768-5002 wherein the pump is adapted for delivering an anti-spastic agent.
5009. The method of any one of items 4768-5002 wherein the pump is adapted for delivering an antibiotic.
5010. The method of any one of items 4768-5002 wherein the pump is adapted for delivering a drug only when changes in the host are detected.
5011. The method of any one of items 4768-5002 wherein the pump is adapted for delivering a drug as a continuous slow release.
5012. The method of any one of items 4768-5002 wherein the pump is adapted for delivering a drug at prescribed dosages in a pulsatile manner.
5013. The method of any one of items 4768-5002 wherein the pump is a programmable drug delivery pump.
5014. The method of any one of items 4768-5002 wherein the pump is adapted for intraocularly delivering a drug.
5015. The method of any one of items 4768-5002 wherein the pump is adapted for intrathecally delivering a drug.
5016. The method of any one of items 4768-5002 wherein the pump is adapted for intraperitoneally delivering a drug.
5017. The method of any one of items 4768-5002 wherein the pump is adapted for intra-arterially delivering a drug.
5018. The method of any one of items 4768-5002 wherein the pump is adapted for intracardiac delivery of a drug. 5019. The method of any one of items 4768-5002 wherein the pump is an implantable osmotic pump.
5020. The method of any one of items 4768-5002 wherein the pump is an ocular drug delivery pump.
5021. The method of any one of items 4768-5002 wherein the pump is metering system.
5022. The method of any one of items 4768-5002 wherein the pump is a peristaltic (roller) pump.
5023. The method of any one of items 4768-5002 wherein the pump is an electronically driven pump.
5024. The method of any one of items 4768-5002 wherein the pump is an elastomeric pump.
5025. The method of any one of items 4768-5002 wherein the pump is a spring contraction pump.
5026. The method of any one of items 4768-5002 wherein the pump is a gas-driven pump.
5027. The method of any one of items 4768-5002 wherein the pump is a hydraulic pump.
5028. The method of any one of items 4768-5002 wherein the pump is a piston-dependent pump.
5029. The method of any one of items 4768-5002 wherein the pump is a non-piston-dependent pump.
5030. The method of any one of items 4768-5002 wherein the pump is a dispensing chamber. 5031. The method of any one of items 4768-5002 wherein the pump is an infusion pump.
5032. The method of any one of items 4768-5002 wherein the pump is a passive pump.
5033. A method for inhibiting scarring comprising placing an implantable insulin pump and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5034. The method of item 5033 wherein the agent is an adensosine A2A receptor antagonist.
5035. The method of item 5033 wherein the agent is an AKT inhibitor.
5036. The method of item 5033 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5037. The method of item 5033 wherein the agent is an alpha 4 integrin antagonist.
5038. The method of item 5033 wherein the agent is an alpha 7 nicotinic receptor agonist.
5039. The method of item 5033 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC)1 neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005
(GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
5040. The method of item 5033 wherein the agent is an apoptosis antagonist.
5041. The method of item 5033 wherein the agent is an apoptosis activator.
5042. The method of item 5033 wherein the agent is a beta 1 integrin antagonist. 5043. The method of item 5033 wherein the agent is a beta tubulin inhibitor.
5044. The method of item 5033 wherein the agent is a blocker of enzyme production in Hepatitis C.
5045. The method of item 5033 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5046. The method of item 5033 wherein the agent is a calcineurin inhibitor.
5047. The method of item 5033 wherein the agent is a caspase 3 inhibitor.
5048. The method of item 5033 wherein the agent is a CC chemokine receptor antagonist.
5049. The method of item 5033 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
5050. The method of item 5033 wherein the agent is a cathepsin B inhibitor.
5051. The method of item 5033 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
5052. The method of item 5033 wherein the agent is a cathepsin L inhibitor.
5053. The method of item 5033 wherein the agent is a CD40 antagonist. 5054. The method of item 5033 wherein the agent is a chemokine receptor agonist.
5055. The method of item 5033 wherein the agent is a chymase inhibitor.
5056. The method of item 5033 wherein the agent is a collagenase antagonist.
5057. The method of item 5033 wherein the agent is a CXCR antagonist.
5058. The method of item 5033 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
5059. The method of item 5033 wherein the agent is a cyclooxygenase 1 inhibitor.
5060. The method of item 5033 wherein the agent is a DHFR inhibitor.
5061. The method of item 5033 wherein the agent is a dual integrin inhibitor.
5062. The method of item 5033 wherein the agent is an elastase inhibitor. 5063. The method of item 5033 wherein the agent is an elongation factor-1 alpha inhibitor.
5064. The method of item 5033 wherein the agent is an endothelial growth factor antagonist.
5065. The method of item 5033 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
5066. The method of item 5033 wherein the agent is an endotoxin antagonist.
5067. The method of item 5033 wherein the agent is an epothilone and tubulin binder.
5068. The method of item 5033 wherein the agent is an estrogen receptor antagonist.
5069. The method of item 5033 wherein the agent is an FGF inhibitor.
5070. The method of item 5033 wherein the agent is a farnexyl transferase inhibitor.
5071. The method of item 5033 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
5072. The method of item 5033 wherein the agent is an FLT-3 kinase inhibitor.
5073. The method of item 5033 wherein the agent is an FGF receptor kinase inhibitor.
5074. The method of item 5033 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201
(Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5075. The method of item 5033 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
5076. The method of item 5033 wherein the agent is a histone deacetylase inhibitor.
5077. The method of item 5033 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 5078. The method of item 5033 wherein the agent is an ICAM inhibitor.
5079. The method of item 5033 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
5080. The method of item 5033 wherein the agent is an IL-2 inhibitor.
5081. The method of item 5033 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPatTT, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5082. The method of item 5033 wherein the agent is an IMPDH (inosine monophosphate). i::jr IC T/ U S O B ./" :L ,17 E IB
5083. The method of item 5033 wherein the agent is an integrin antagonist.
5084. The method of item 5033 wherein the agent is an interleukin antagonist.
5 5085. The method of item 5033 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
5086. The method of item 5033 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5087. The method of item 5033 wherein the agent is an 10 isozyme selective delta protein kinase C inhibitor.
5088. The method of item 5033 wherein the agent a JAK3 enzyme inhibitor.
5089. The method of item 5033 wherein the agent is a JNK inhibitor.
15 5090. The method of item 5033 wherein the agent is a kinase inhibitor.
5091. The method of item 5033 wherein the agent is kinesin antagonist.
5092. The method of item 5033 wherein the agent is a kinesin 20 antagonist.
5093. The method of item 5033 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi
25 Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 CT./USOe./',i :|L7l5iB
and 2728 (CAS No.D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS 5 No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) 10 (Pfizer), and analogue or derivative thereof.
5094. The method of item 5033 wherein the agent is an MAP kinase inhibitor.
5095. The method of item 5033 wherein the agent is a matrix metalloproteinase inhibitor.
15 5096. The method of item 5033 wherein the agent is an MCP-
CCR2 inhibitor.
5097. The method of item 5033 wherein the agent is an mTOR inhibitor.
5098. The method of item 5033 wherein the agent is an mTOR 20 kinase inhibitor.
5099. The method of item 5033 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson,
25 DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
5100. The method of item 5033 wherein the agent is an MIF inhibitor.
5101. The method of item 5033 wherein the agent is an MMP inhibitor.
5102. The method of item 5033 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
5103. The method of item 5033 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 5104. The method of item 5033 wherein the agent is a nitric oxide agonist.
5105. The method of item 5033 wherein the agent is an ornithine decarboxylase inhibitor.
5106. The method of item 5033 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5107. The method of item 5033 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
5108. The method of item 5033 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
5109. The method of item 5033 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKIine), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
5110. The method of item 5033 wherein the agent is a phosphatase inhibitor.
5111. The method of item 5033 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5112. The method of item 5033 wherein the agent is a PKC inhibitor.
5113. The method of item 5033 wherein the agent is a platelet activating factor antagonist.
5114. The method of item 5033 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
5115. The method of item 5033 wherein the agent is a prolyl hydroxylase inhibitor.
5116. The method of item 5033 wherein the agent is a polymorphonuclear neutrophil inhibitor.
5117. The method of item 5033 wherein the agent is a protein kinase B inhibitor.
5118. The method of item 5033 wherein the agent is a protein kinase C stimulant.
5119. The method of item 5033 wherein the agent is a purine nucleoside analogue.
5120. The method of item 5033 wherein the agent is a purinoreceptor P2X antagonist.
5121. The method of item 5033 wherein the agent is a Raf kinase inhibitor. 5122. The method of item 5033 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
5123. The method of item 5033 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5124. The method of item 5033 wherein the agent is an SDF-1 antagonist.
5125. The method of item 5033 wherein the agent is a sheddase inhibitor.
5126. The method of item 5033 wherein the agent is an SRC inhibitor.
5127. The method of item 5033 wherein the agent is a stromelysin inhibitor.
5128. The method of item 5033 wherein the agent is an Syk kinase inhibitor.
5129. The method of item 5033 wherein the agent is a telomerase inhibitor.
5130. The method of item 5033 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5131. The method of item 5033 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certoiizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YS1L6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5132. The method of item 5033 wherein the agent is a Toll receptor inhibitor.
5133. The method of item 5033 wherein the agent is a tubulin antagonist.
5134. The method of item 5033 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paraceisian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
5135. The method of item 5033 wherein the agent is a VEGF inhibitor.
5136. The method of item 5033 wherein the agent is a vitamin D receptor agonist.
5137. The method of item 5033 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
5138. The method of item 5033 wherein the agent is AP-23573 (an mTOR inhibitor). 5139. The method of item 5033 wherein the agent is synthadotin (a tubulin antagonist).
5140. The method of item 5033 wherein the agent is S-0885 (a collagenase inhibitor).
5141. The method of item 5033 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
5142. The method of item 5033 wherein the agent is ixabepilone (an epithilone).
5143. The method of item 5033 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
5144. The method of item 5033 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
5145. The method of item 5033 wherein the agent is ABT-518 (an angiogenesis inhibitor).
5146. The method of item 5033 wherein the agent is combretastatin (an angiogenesis inhibitor).
5147. The method of item 5033 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
5148. The method of item 5033 wherein the agent is SB- 715992 (a kinesin antagonist).
5149. The method of item 5033 wherein the agent is temsirolimus (an mTOR inhibitor).
5150. The method of item 5033 wherein the agent is adalimumab (a TNFα antagonist). 5151. The method of item 5033, wherein the composition comprises a polymer.
5152. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
5153. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
5154. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
5155. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
5156. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
5157. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
5158. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
5159. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
5160. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 5161. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
5162. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
5163. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
5164. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
5165. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
5166. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
5167. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
5168. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
5169. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
5170. The method of item 5033, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
5171. The method of item 5033, wherein the composition further comprises a second pharmaceutically active agent. 5172. The method of item 5033, wherein the composition further comprises an anti-inflammatory agent.
5173. The method of item 5033, wherein the composition further comprises an agent that inhibits infection.
5174. The method of item 5033, wherein the composition further comprises an anthracycline.
5175. The method of item 5033, wherein the composition further comprises doxorubicin.
5176. The method of item 5033 wherein the composition further comprises mitoxantrone.
5177. The method of item 5033 wherein the composition further comprises a fluoropyrimidine.
5178. The method of item 5033, wherein the composition further comprises 5-fluorouracil (5-FU).
5179. The method of item 5033, wherein the composition further comprises a folic acid antagonist.
5180. The method of item 5033, wherein the composition further comprises methotrexate.
5181. The method of item 5033, wherein the composition further comprises a podophylotoxin.
5182. The method of item 5033, wherein the composition further comprises etoposide.
5183. The method of item 5033, wherein the composition further comprises camptothecin. 5184. The method of item 5033, wherein the composition further comprises a hydroxyurea.
5185. The method of item 5033, wherein the composition further comprises a platinum complex.
5186. The method of item 5033, wherein the composition further comprises cisplatin.
5187. The method of item 5033 wherein the composition further comprises an anti-thrombotic agent.
5188. The method of item 5033, wherein the composition further comprises a visualization agent.
5189. The method of item 5033, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
5190. The method of item 5033, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
5191. The method of item 5033, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
5192. The method of item 5033, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
5193. The method of item 5033, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 5194. The method of item 5033, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
5195. The method of item 5033, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
5196. The method of item 5033 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
5197. The method of item 5033 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
5198. The method of item 5033 wherein the composition further comprises an inflammatory cytokine.
5199. The method of item 5033 wherein the composition further comprises an agent that stimulates cell proliferation.
5200. The method of item 5033 wherein the composition further comprises a polymeric carrier.
5201. The method of item 5033 wherein the composition is in the form of a gel, paste, or spray.
5202. The method of item 5033 wherein the sensor is partially constructed with the agent or the composition.
5203. The method of item 5033 wherein the sensor is impregnated with the agent or the composition. 5204. The method of item 5033, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
5205. The method of item 5033, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
5206. The method of item 5033 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
5207. The method of item 5033, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
5208. The method of item 5033 wherein the agent or the composition is located within pores or holes of the sensor.
5209. The method of item 5033 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
5210. The method of item 5033 wherein the sensor further comprises an echogenic material.
5211. The method of item 5033 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
5212. The method of item 5033 wherein the sensor is sterile.
5213. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
5214. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue. 5215. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
5216. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
5217. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
5218. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
5219. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
5220. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
5221. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
5222. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate. 5223. The method of item 5033 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
5224. The method of item 5033 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
5225. The method of item 5033 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
5226. The method of item 5033 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
5227. The method of item 5033 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
5228. The method of item 5033 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
5229. The method of item 5033 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
5230. The method of item 5033 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied. 5231. The method of item 5033 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
5232. The method of item 5033 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
5233. The method of item 5033 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
5234. The method of item 5033 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
5235. The method of item 5033, wherein the sensor further comprises a coating, and the coating is a uniform coating.
5236. The method of item 5033, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
5237. The method of item 5033, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
5238. The method of item 5033, wherein the sensor further comprises a coating, and the coating is a patterned coating.
5239. The method of item 5033, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less. 5240. The method of item 5033, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
5241. The method of item 5033, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
5242. The method of item 5033, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
5243. The method of item 5033, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
5244. The method of item 5033, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5245. The method of item 5033, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5246. The method of item 5033, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
5247. The method of item 5033, wherein the sensor further comprises a coating, and the coating comprises a polymer.
5248. The method of item 5033, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition. 5249. The method of item 5033, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
5250. The method of item 5033 wherein the agent or the composition is affixed to the sensor.
5251. The method of item 5033 wherein the agent or the composition is covalently attached to the sensor.
5252. The method of item 5033 wherein the agent or the composition is non-covalently attached to the sensor.
5253. The method of item 5033 wherein the sensor comprises a coating that absorbs the agent or the composition.
5254. The method of item 5033 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
5255. The method of item 5033 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
5256. The method of item 5033 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
5257. The method of item 5033 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
5258. The method of item 5033 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
5259. The method of item 5033 wherein the sensor is linked to a pump. 5260. The method of item 5033 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
5261. The method of item 5033 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
5262. The method of item 5033 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
5263. The method of item 5033 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
5264. The method of item 5033 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
5265. The method of item 5033 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
5266. The method of item 5033 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
5267. The method of item 5033 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
5268. The method of any one of items 5033-5267 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense mediation through the catheter.
5269. A method for inhibiting scarring comprising placing an intrathecal drug delivery pump and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5270. The method of item 5269 wherein the agent is an adensosine A2A receptor antagonist.
5271. The method of item 5269 wherein the agent is an AKT inhibitor.
5272. The method of item 5269 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5273. The method of item 5269 wherein the agent is an alpha 4 integrin antagonist.
5274. The method of item 5269 wherein the agent is an alpha 7 nicotinic receptor agonist.
5275. The method of item 5269 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), lDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT- 116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
5276. The method of item 5269 wherein the agent is an apoptosis antagonist.
5277. The method of item 5269 wherein the agent is an apoptosis activator.
5278. The method of item 5269 wherein the agent is a beta 1 integrin antagonist.
5279. The method of item 5269 wherein the agent is a beta tubulin inhibitor. 5280. The method of item 5269 wherein the agent is a blocker of enzyme production in Hepatitis C.
5281. The method of item 5269 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5282. The method of item 5269 wherein the agent is a calcineurin inhibitor.
5283. The method of item 5269 wherein the agent is a caspase 3 inhibitor.
5284. The method of item 5269 wherein the agent is a CC chemokine receptor antagonist.
5285. The method of item 5269 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
5286. The method of item 5269 wherein the agent is a cathepsin B inhibitor.
5287. The method of item 5269 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
5288. The method of item 5269 wherein the agent is a cathepsin L inhibitor.
5289. The method of item 5269 wherein the agent is a CD40 antagonist.
5290. The method of item 5269 wherein the agent is a chemokine receptor agonist. 5291. The method of item 5269 wherein the agent is a chymase inhibitor.
5292. The method of item 5269 wherein the agent is a collagenase antagonist.
5293. The method of item 5269 wherein the agent is a CXCR antagonist.
5294. The method of item 5269 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
5295. The method of item 5269 wherein the agent is a cyclooxygenase 1 inhibitor.
5296. The method of item 5269 wherein the agent is a DHFR inhibitor.
5297. The method of item 5269 wherein the agent is a dual integrin inhibitor.
5298. The method of item 5269 wherein the agent is an elastase inhibitor.
5299. The method of item 5269 wherein the agent is an elongation factor-1 alpha inhibitor. 5300. The method of item 5269 wherein the agent is an endothelial growth factor antagonist.
5301. The method of item 5269 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
5302. The method of item 5269 wherein the agent is an endotoxin antagonist.
5303. The method of item 5269 wherein the agent is an epothilone and tubulin binder.
5304. The method of item 5269 wherein the agent is an estrogen receptor antagonist.
5305. The method of item 5269 wherein the agent is an FGF inhibitor.
5306. The method of item 5269 wherein the agent is a farnexyl transferase inhibitor.
5307. The method of item 5269 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
5308. The method of item 5269 wherein the agent is an FLT-3 kinase inhibitor.
5309. The method of item 5269 wherein the agent is an FGF receptor kinase inhibitor.
5310. The method of item 5269 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11 -9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5311. The method of item 5269 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
5312. The method of item 5269 wherein the agent is a histone deacetylase inhibitor.
5313. The method of item 5269 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 5314. The method of item 5269 wherein the agent is an ICAM inhibitor.
5315. The method of item 5269 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
5316. The method of item 5269 wherein the agent is an IL-2 inhibitor.
5317. The method of item 5269 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from Merϋon Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5318. The method of item 5269 wherein the agent is an IMPDH (inosine monophosphate). 5319. The method of item 5269 wherein the agent is an integrin antagonist.
5320. The method of item 5269 wherein the agent is an interleukin antagonist.
5321. The method of item 5269 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
5322. The method of item 5269 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5323. The method of item 5269 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
5324. The method of item 5269 wherein the agent a JAK3 enzyme inhibitor.
5325. The method of item 5269 wherein the agent is a JNK inhibitor.
5326. The method of item 5269 wherein the agent is a kinase inhibitor.
5327. The method of item 5269 wherein the agent is kinesin antagonist.
5328. The method of item 5269 wherein the agent is a kinesin antagonist.
5329. The method of item 5269 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
5330. The method of item 5269 wherein the agent is an MAP kinase inhibitor.
5331. The method of item 5269 wherein the agent is a matrix metalloproteinase inhibitor.
5332. The method of item 5269 wherein the agent is an MCP-
CCR2 inhibitor.
5333. The method of item 5269 wherein the agent is an mTOR inhibitor.
5334. The method of item 5269 wherein the agent is an mTOR kinase inhibitor.
5335. The method of item 5269 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
5336. The method of item 5269 wherein the agent is an MIF inhibitor.
5337. The method of item 5269 wherein the agent is an MMP inhibitor.
5338. The method of item 5269 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
5339. The method of item 5269 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 5340. The method of item 5269 wherein the agent is a nitric oxide agonist.
5341. The method of item 5269 wherein the agent is an ornithine decarboxylase inhibitor.
5342. The method of item 5269 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5343. The method of item 5269 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
5344. The method of item 5269 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
5345. The method of item 5269 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1 ), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
5346. The method of item 5269 wherein the agent is a phosphatase inhibitor.
5347. The method of item 5269 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5348. The method of item 5269 wherein the agent is a PKC inhibitor.
5349. The method of item 5269 wherein the agent is a platelet activating factor antagonist.
5350. The method of item 5269 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
5351. The method of item 5269 wherein the agent is a prolyl hydroxylase inhibitor.
5352. The method of item 5269 wherein the agent is a polymorphonuclear neutrophil inhibitor.
5353. The method of item 5269 wherein the agent is a protein kinase B inhibitor.
5354. The method of item 5269 wherein the agent is a protein kinase C stimulant.
5355. The method of item 5269 wherein the agent is a purine nucleoside analogue.
5356. The method of item 5269 wherein the agent is a purinoreceptor P2X antagonist.
5357. The method of item 5269 wherein the agent is a Raf kinase inhibitor. 5358. The method of item 5269 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
5359. The method of item 5269 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5360. The method of item 5269 wherein the agent is an SDF-1 antagonist.
5361. The method of item 5269 wherein the agent is a sheddase inhibitor.
5362. The method of item 5269 wherein the agent is an SRC inhibitor.
5363. The method of item 5269 wherein the agent is a stromelysin inhibitor.
5364. The method of item 5269 wherein the agent is an Syk kinase inhibitor.
5365. The method of item 5269 wherein the agent is a telomerase inhibitor.
5366. The method of item 5269 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5367. The method of item 5269 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPFM 32294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5368. The method of item 5269 wherein the agent is a Toll receptor inhibitor.
5369. The method of item 5269 wherein the agent is a tubulin antagonist.
5370. The method of item 5269 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosyiate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
5371. The method of item 5269 wherein the agent is a VEGF inhibitor.
5372. The method of item 5269 wherein the agent is a vitamin D receptor agonist.
5373. The method of item 5269 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
5374. The method of item 5269 wherein the agent is AP-23573 (an mTOR inhibitor). 5375. The method of item 5269 wherein the agent is synthadotin (a tubulin antagonist).
5376. The method of item 5269 wherein the agent is S-0885 (a collagenase inhibitor).
5377. The method of item 5269 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
5378. The method of item 5269 wherein the agent is ixabepilone (an epithilone).
5379. The method of item 5269 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
5380. The method of item 5269 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
5381. The method of item 5269 wherein the agent is ABT-518 (an angiogenesis inhibitor).
5382. The method of item 5269 wherein the agent is combretastatin (an angiogenesis inhibitor).
5383. The method of item 5269 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
5384. The method of item 5269 wherein the agent is SB- 715992 (a kinesin antagonist).
5385. The method of item 5269 wherein the agent is temsirolimus (an mTOR inhibitor).
5386. The method of item 5269 wherein the agent is adalimumab (a TNFα antagonist). 5387. The method of item 5269, wherein the composition comprises a polymer.
5388. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
5389. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
5390. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
5391. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
5392. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
5393. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
5394. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
5395. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
5396. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 5397. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
5398. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
5399. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
5400. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
5401. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
5402. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
5403. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
5404. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
5405. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
5406. The method of item 5269, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
5407. The method of item 5269, wherein the composition further comprises a second pharmaceutically active agent. 5408. The method of item 5269, wherein the composition further comprises an anti-inflammatory agent.
5409. The method of item 5269, wherein the composition further comprises an agent that inhibits infection.
5410. The method of item 5269, wherein the composition further comprises an anthracycline.
5411. The method of item 5269, wherein the composition further comprises doxorubicin.
5412. The method of item 5269 wherein the composition further comprises mitoxantrone.
5413. The method of item 5269 wherein the composition further comprises a fluoropyrimidine.
5414. The method of item 5269, wherein the composition further comprises 5-fluorouracil (5-FU).
5415. The method of item 5269, wherein the composition further comprises a folic acid antagonist.
5416. The method of item 5269, wherein the composition further comprises methotrexate.
5417. The method of item 5269, wherein the composition further comprises a podophylotoxin.
5418. The method of item 5269, wherein the composition further comprises etoposide.
5419. The method of item 5269, wherein the composition further comprises camptothecin. 5420. The method of item 5269, wherein the composition further comprises a hydroxyurea.
5421. The method of item 5269, wherein the composition further comprises a platinum complex.
5422. The method of item 5269, wherein the composition further comprises cisplatin.
5423. The method of item 5269 wherein the composition further comprises an anti-thrombotic agent.
5424. The method of item 5269, wherein the composition further comprises a visualization agent.
5425. The method of item 5269, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
5426. The method of item 5269, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
5427. The method of item 5269, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
5428. The method of item 5269, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
5429. The method of item 5269, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 5430. The method of item 5269, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
5431. The method of item 5269, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
5432. The method of item 5269 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
5433. The method of item 5269 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
5434. The method of item 5269 wherein the composition further comprises an inflammatory cytokine.
5435. The method of item 5269 wherein the composition further comprises an agent that stimulates cell proliferation.
5436. The method of item 5269 wherein the composition further comprises a polymeric carrier.
5437. The method of item 5269 wherein the composition is in the form of a gel, paste, or spray.
5438. The method of item 5269 wherein the sensor is partially constructed with the agent or the composition.
5439. The method of item 5269 wherein the sensor is impregnated with the agent or the composition. 5440. The method of item 5269, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
5441. The method of item 5269, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
5442. The method of item 5269 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
5443. The method of item 5269, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
5444. The method of item 5269 wherein the agent or the composition is located within pores or holes of the sensor.
5445. The method of item 5269 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
5446. The method of item 5269 wherein the sensor further comprises an echogenic material.
5447. The method of item 5269 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
5448. The method of item 5269 wherein the sensor is sterile.
5449. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
5450. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue. 5451. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
5452. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
5453. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
5454. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
5455. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
5456. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
5457. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
5458. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate. 5459. The method of item 5269 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
5460. The method of item 5269 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
5461. The method of item 5269 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
5462. The method of item 5269 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
5463. The method of item 5269 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
5464. The method of item 5269 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
5465. The method of item 5269 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
5466. The method of item 5269 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied. 5467. The method of item 5269 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
5468. The method of item 5269 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
5469. The method of item 5269 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
5470. The method of item 5269 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
5471. The method of item 5269, wherein the sensor further comprises a coating, and the coating is a uniform coating.
5472. The method of item 5269, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
5473. The method of item 5269, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
5474. The method of item 5269, wherein the sensor further comprises a coating, and the coating is a patterned coating.
5475. The method of item 5269, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less. 5476. The method of item 5269, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
5477. The method of item 5269, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
5478. The method of item 5269, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
5479. The method of item 5269, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
5480. The method of item 5269, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5481. The method of item 5269, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5482. The method of item 5269, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
5483. The method of item 5269, wherein the sensor further comprises a coating, and the coating comprises a polymer.
5484. The method of item 5269, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition. 5485. The method of item 5269, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
5486. The method of item 5269 wherein the agent or the composition is affixed to the sensor.
5487. The method of item 5269 wherein the agent or the composition is covalently attached to the sensor.
5488. The method of item 5269 wherein the agent or the composition is non-covalently attached to the sensor.
5489. The method of item 5269 wherein the sensor comprises a coating that absorbs the agent or the composition.
5490. The method of item 5269 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
5491. The method of item 5269 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
5492. The method of item 5269 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
5493. The method of item 5269 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
5494. The method of item 5269 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
5495. The method of item 5269 wherein the sensor is linked to a pump. 5496. The method of item 5269 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
5497. The method of item 5269 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
5498. The method of item 5269 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
5499. The method of item 5269 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
5500. The method of item 5269 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
5501. The method of item 5269 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
5502. The method of item 5269 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
5503. The method of item 5269 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
5504. The method of any one of items 5269-5503 wherein the pump is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord. 5505. The method of any one of items 5269-5503 wherein the pump is adapted for delivering a drug to the brain.
5506. The method of any one of items 5269-5503 wherein the pump is adapted for intrathecal delivering baclofen.
5507. The method of any one of items 5269-5503 wherein the pump further comprises an intraspinal catheter.
5508. The method of any one of items 5269-5503 further comprising a second intrathecal drug delivery pump.
5509. The method of any one of items 5269-5503 wherein the pump further comprises a catheter and an electrode.
5510. A method for inhibiting scarring comprising placing an implantable drug delivery pump for chemotherapy and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5511. The method of item 5510 wherein the agent is an adensosine A2A receptor antagonist.
5512. The method of item 5510 wherein the agent is an AKT inhibitor.
5513. The method of item 5510 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects
No. 5754 (Merck KgaA).
5514. The method of item 5510 wherein the agent is an alpha 4 integrin antagonist.
5515. The method of item 5510 wherein the agent is an alpha 7 nicotinic receptor agonist. 5516. The method of item 5510 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium. (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 5517. The method of item 5510 wherein the agent is an apoptosis antagonist.
5518. The method of item 5510 wherein the agent is an apoptosis activator.
5519. The method of item 5510 wherein the agent is a beta 1 integrin antagonist.
( 5520. The method of item 5510 wherein the agent is a beta tubulin inhibitor.
5521. The method of item 5510 wherein the agent is a blocker of enzyme production in Hepatitis C.
5522. The method of item 5510 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5523. The method of item 5510 wherein the agent is a calcineurin inhibitor.
5524. The method of item 5510 wherein the agent is a caspase
3 inhibitor.
5525. The method of item 5510 wherein the agent is a CC chemokine receptor antagonist.
5526. The method of item 5510 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin,
KRX-0403, and an analogue or derivative thereof.
5527. The method of item 5510 wherein the agent is a cathepsin B inhibitor.
5528. The method of item 5510 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), 1NPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
5529. The method of item 5510 wherein the agent is a cathepsin L inhibitor.
5530. The method of item 5510 wherein the agent is a CD40 antagonist.
5531. The method of item 5510 wherein the agent is a chemokine receptor agonist.
5532. The method of item 5510 wherein the agent is a chymase inhibitor.
5533. The method of item 5510 wherein the agent is a collagenase antagonist.
5534. The method of item 5510 wherein the agent is a CXCR antagonist.
5535. The method of item 5510 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
5536. The method of item 5510 wherein the agent is a cyclooxygenase 1 inhibitor. 5537. The method of item 5510 wherein the agent is a DHFR inhibitor.
5538. The method of item 5510 wherein the agent is a dual integrin inhibitor.
5539. The method of item 5510 wherein the agent is an elastase inhibitor.
5540. The method of item 5510 wherein the agent is an elongation factor-1 alpha inhibitor.
5541. The method of item 5510 wherein the agent is an endothelial growth factor antagonist.
5542. The method of item 5510 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
5543. The method of item 5510 wherein the agent is an endotoxin antagonist.
5544. The method of item 5510 wherein the agent is an epothilone and tubulin binder.
5545. The method of item 5510 wherein the agent is an estrogen receptor antagonist. 5546. The method of item 5510 wherein the agent is an FGF inhibitor.
5547. The method of item 5510 wherein the agent is a famexyl transferase inhibitor.
5548. The method of item 5510 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
5549. The method of item 5510 wherein the agent is an FLT-3 kinase inhibitor.
5550. The method of item 5510 wherein the agent is an FGF receptor kinase inhibitor.
5551. The method of item 5510 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5552. The method of item 5510 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof. 5553. The method of item 5510 wherein the agent is a histone deacetylase inhibitor.
5554. The method of item 5510 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
5555. The method of item 5510 wherein the agent is an ICAM inhibitor.
5556. The method of item 5510 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
5557. The method of item 5510 wherein the agent is an IL-2 inhibitor.
5558. The method of item 5510 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5559. The method of item 5510 wherein the agent is an IMPDH (inosine monophosphate).
5560. The method of item 5510 wherein the agent is an integrin antagonist.
5561. The method of item 5510 wherein the agent is an interleukin antagonist.
5562. The method of item 5510 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
5563. The method of item 5510 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5564. The method of item 5510 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
5565. The method of item 5510 wherein the agent a JAK3 enzyme inhibitor.
5566. The method of item 5510 wherein the agent is a JNK inhibitor.
5567. The method of item 5510 wherein the agent is a kinase inhibitor.
5568. The method of item 5510 wherein the agent is kinesin antagonist. 5569. The method of item 5510 wherein the agent is a kinesin antagonist.
5570. The method of item 5510 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No.D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
5571. The method of item 5510 wherein the agent is an MAP kinase inhibitor.
5572. The method of item 5510 wherein the agent is a matrix metalloproteinase inhibitor.
5573. The method of item 5510 wherein the agent is an MCP- CCR2 inhibitor.
5574. The method of item 5510 wherein the agent is an mTOR inhibitor.
5575. The method of item 5510 wherein the agent is an mTOR kinase inhibitor. 5576. The method of item 5510 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
5577. The method of item 5510 wherein the agent is an MIF inhibitor.
5578. The method of item 5510 wherein the agent is an MMP inhibitor.
5579. The method of item 5510 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
5580. The method of item 5510 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
5581. The method of item 5510 wherein the agent is a nitric oxide agonist.
5582. The method of item 5510 wherein the agent is an ornithine decarboxylase inhibitor.
5583. The method of item 5510 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5584. The method of item 5510 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
5585. The method of item 5510 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 5586. The method of item 5510 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
5587. The method of item 5510 wherein the agent is a phosphatase inhibitor.
5588. The method of item 5510 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPFM 17658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5589. The method of item 5510 wherein the agent is a PKC inhibitor.
5590. The method of item 5510 wherein the agent is a platelet activating factor antagonist.
5591. The method of item 5510 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
5592. The method of item 5510 wherein the agent is a prolyl hydroxylase inhibitor.
5593. The method of item 5510 wherein the agent is a polymorphonuclear neutrophil inhibitor.
5594. The method of item 5510 wherein the agent is a protein kinase B inhibitor.
5595. The method of item 5510 wherein the agent is a protein kinase C stimulant. 5596. The method of item 5510 wherein the agent is a purine nucleoside analogue.
5597. The method of item 5510 wherein the agent is a purinoreceptor P2X antagonist.
5598. The method of item 5510 wherein the agent is a Raf kinase inhibitor.
5599. The method of item 5510 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
5600. The method of item 5510 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5601. The method of item 5510 wherein the agent is an SDF-1 antagonist.
5602. The method of item 5510 wherein the agent is a sheddase inhibitor.
5603. The method of item 5510 wherein the agent is an SRC inhibitor.
5604. The method of item 5510 wherein the agent is a stromelysin inhibitor.
5605. The method of item 5510 wherein the agent is an Syk kinase inhibitor.
5606. The method of item 5510 wherein the agent is a telomerase inhibitor.
5607. The method of item 5510 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5608. The method of item 5510 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5609. The method of item 5510 wherein the agent is a Toll receptor inhibitor.
5610. The method of item 5510 wherein the agent is a tubulin antagonist.
5611. The method of item 5510 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), 2D-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
5612. The method of item 5510 wherein the agent is a VEGF inhibitor. 5613. The method of item 5510 wherein the agent is a vitamin D receptor agonist.
5614. The method of item 5510 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
5615. The method of item 5510 wherein the agent is AP-23573
(an mTOR inhibitor).
5616. The method of item 5510 wherein the agent is synthadotin (a tubulin antagonist).
5617. The method of item 5510 wherein the agent is S-0885 (a coliagenase inhibitor).
5618. The method of item 5510 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
5619. The method of item 5510 wherein the agent is ixabepilone (an epithilone).
5620. The method of item 5510 wherein the agent is IDN-5390
(an angiogenesis inhibitor).
5621. The method of item 5510 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
5622. The method of item 5510 wherein the agent is ABT-518 (an angiogenesis inhibitor).
5623. The method of item 5510 wherein the agent is combretastatin (an angiogenesis inhibitor).
5624. The method of item 5510 wherein the agent is anecortave acetate (an angiogenesis inhibitor). 5625. The method of item 5510 wherein the agent is SB- 715992 (a kinesin antagonist).
5626. The method of item 5510 wherein the agent is temsirolimus (an mTOR inhibitor).
5627. The method of item 5510 wherein the agent is adalimumab (a TNFα antagonist).
5628. The method of item 5510, wherein the composition comprises a polymer.
5629. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
5630. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
5631. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
5632. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
5633. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
5634. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
5635. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 5636. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
5637. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
5638. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
5639. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
5640. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
5641. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
5642. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
5643. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
5644. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
5645. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer. 5646. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
5647. The method of item 5510, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
5648. The method of item 5510, wherein the composition further comprises a second pharmaceutically active agent.
5649. The method of item 5510, wherein the composition further comprises an anti-inflammatory agent.
5650. The method of item 5510, wherein the composition further comprises an agent that inhibits infection.
5651. The method of item 5510, wherein the composition further comprises an anthracycline.
5652. The method of item 5510, wherein the composition further comprises doxorubicin.
5653. The method of item 5510 wherein the composition further comprises mitoxantrone.
5654. The method of item 5510 wherein the composition further comprises a fluoropyrimidine.
5655. The method of item 5510, wherein the composition further comprises 5-fluorouracil (5-FU).
5656. The method of item 5510, wherein the composition further comprises a folic acid antagonist.
5657. The method of item 5510, wherein the composition further comprises methotrexate. 5658. The method of item 5510, wherein the composition further comprises a podophylotoxin.
5659. The method of item 5510, wherein the composition further comprises etoposide.
5660. The method of item 5510, wherein the composition further comprises camptothecin.
5661. The method of item 5510, wherein the composition further comprises a hydroxyurea.
5662. The method of item 5510, wherein the composition further comprises a platinum complex.
5663. The method of item 5510, wherein the composition further comprises cisplatin.
5664. The method of item 5510 wherein the composition further comprises an anti-thrombotic agent.
5665. The method of item 5510, wherein the composition further comprises a visualization agent.
5666. The method of item 5510, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
5667. The method of item 5510, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
5668. The method of item 5510, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material. 5669. The method of item 5510, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
5670. The method of item 5510, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
5671. The method of item 5510, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
5672. The method of item 5510, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
5673. The method of item 5510 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
5674. The method of item 5510 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
5675. The method of item 5510 wherein the composition further comprises an inflammatory cytokine.
5676. The method of item 5510 wherein the composition further comprises an agent that stimulates cell proliferation.
5677. The method of item 5510 wherein the composition further comprises a polymeric carrier. 5678. The method of item 5510 wherein the composition is in the form of a gel, paste, or spray.
5679. The method of item 5510 wherein the sensor is partially constructed with the agent or the composition.
5680. The method of item 5510 wherein the sensor is impregnated with the agent or the composition.
5681. The method of item 5510, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
5682. The method of item 5510, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
5683. The method of item 5510 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
5684. The method of item 5510, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
5685. The method of item 5510 wherein the agent or the composition is located within pores or holes of the sensor.
5686. The method of item 5510 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
5687. The method of item 5510 wherein the sensor further comprises an echogenic material.
5688. The method of item 5510 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
5689. The method of item 5510 wherein the sensor is sterile. 5690. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
5691. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
5692. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
5693. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
5694. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
5695. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
5696. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
5697. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days. 5698. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
5699. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
5700. The method of item 5510 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
5701. The method of item 5510 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
5702. The method of item 5510 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
5703. The method of item 5510 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
5704. The method of item 5510 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
5705. The method of item 5510 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
5706. The method of item 5510 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied. 5707. The method of item 5510 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
5708. The method of item 5510 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
5709. The method of item 5510 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
5710. The method of item 5510 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
5711. The method of item 5510 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
5712. The method of item 5510, wherein the sensor further comprises a coating, and the coating is a uniform coating.
5713. The method of item 5510, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
5714. The method of item 5510, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
5715. The method of item 5510, wherein the sensor further comprises a coating, and the coating is a patterned coating. 5716. The method of item 5510, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
5717. The method of item 5510, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
5718. The method of item 5510, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
5719. The method of item 5510, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
5720. The method of item 5510, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
5721. The method of item 5510, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
5722. The method of item 5510, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5723. The method of item 5510, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
5724. The method of item 5510, wherein the sensor further comprises a coating, and the coating comprises a polymer. 5725. The method of item 5510, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
5726. The method of item 5510, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
5727. The method of item 5510 wherein the agent or the composition is affixed to the sensor.
5728. The method of item 5510 wherein the agent or the composition is covalently attached to the sensor.
5729. The method of item 5510 wherein the agent or the composition is non-covalently attached to the sensor.
5730. The method of item 5510 wherein the sensor comprises a coating that absorbs the agent or the composition.
5731. The method of item 5510 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
5732. The method of item 5510 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
5733. The method of item 5510 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
5734. The method of item 5510 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
5735. The method of item 5510 wherein the sensor is completely covered with a mesh that contains the agent or the composition. 5736. The method of item 5510 wherein the sensor is linked to a pump.
5737. The method of item 5510 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
5738. The method of item 5510 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
5739. The method of item 5510 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
5740. The method of item 5510 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
5741. The method of item 5510 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
5742. The method of item 5510 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
5743. The method of item 5510 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
5744. The method of item 5510 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor. 5745. The method of any one of items 5510-5744 wherein the pump is adapted for delivering 2'-deoxy 5-fluorouridine.
5746. The method of any one of items 5510-5744 wherein the host has a solid tumor, and the pump is adapted for infusing a chemotherapeutic agent to the solid tumor.
5747. The method of any one of items 5510-5744 wherein the host has a tumor, and the pump is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
5748. The method of any one of items 5510-5744 wherein the host has a hepatic tumor, and the pump is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host.
5749. A method for inhibiting scarring comprising placing a drug delivery pump for treating heart disease and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5750. The method of item 5749 wherein the agent is an adensosine A2A receptor antagonist.
5751. The method of item 5749 wherein the agent is an AKT inhibitor.
5752. The method of item 5749 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5753. The method of item 5749 wherein the agent is an alpha 4 integrin antagonist. 5754. The method of item 5749 wherein the agent is an alpha 7 nicotinic receptor agonist.
5755. The method of item 5749 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEΞAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
5756. The method of item 5749 wherein the agent is an apoptosis antagonist.
5757. The method of item 5749 wherein the agent is an apoptosis activator.
5758. The method of item 5749 wherein the agent is a beta 1 integrin antagonist.
5759. The method of item 5749 wherein the agent is a beta tubulin inhibitor.
5760. The method of item 5749 wherein the agent is a blocker of enzyme production in Hepatitis C.
5761. The method of item 5749 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5762. The method of item 5749 wherein the agent is a calcineurin inhibitor.
5763. The method of item 5749 wherein the agent is a caspase 3 inhibitor.
5764. The method of item 5749 wherein the agent is a CC chemokine receptor antagonist.
5765. The method of item 5749 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
5766. The method of item 5749 wherein the agent is a cathepsin B inhibitor. 5767. The method of item 5749 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
5768. The method of item 5749 wherein the agent is a cathepsin L inhibitor.
5769. The method of item 5749 wherein the agent is a CD40 antagonist.
5770. The method of item 5749 wherein the agent is a chemokine receptor agonist.
5771. The method of item 5749 wherein the agent is a chymase inhibitor.
5772. The method of item 5749 wherein the agent is a collagenase antagonist.
5773. The method of item 5749 wherein the agent is a CXCR antagonist.
5774. The method of item 5749 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 5775. The method of item 5749 wherein the agent is a cyclooxygenase 1 inhibitor.
5776. The method of item 5749 wherein the agent is a DHFR inhibitor.
5777. The method of item 5749 wherein the agent is a dual integrin inhibitor.
5778. The method of item 5749 wherein the agent is an eiastase inhibitor.
5779. The method of item 5749 wherein the agent is an elongation factor-1 alpha inhibitor.
5780. The method of item 5749 wherein the agent is an endothelial growth factor antagonist.
5781. The method of item 5749 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
5782. The method of item 5749 wherein the agent is an endotoxin antagonist.
5783. The method of item 5749 wherein the agent is an epothilone and tubulin binder. 5784. The method of item 5749 wherein the agent is an estrogen receptor antagonist.
5785. The method of item 5749 wherein the agent is an FGF inhibitor.
5786. The method of item 5749 wherein the agent is a famexyl transferase inhibitor.
5787. The method of item 5749 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme),
Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
5788. The method of item 5749 wherein the agent is an FLT-3 kinase inhibitor.
5789. The method of item 5749 wherein the agent is an FGF receptor kinase inhibitor.
5790. The method of item 5749 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11 -9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5791. The method of item 5749 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehyclro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
5792. The method of item 5749 wherein the agent is a histone deacetylase inhibitor.
5793. The method of item 5749 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
5794. The method of item 5749 wherein the agent is an ICAM inhibitor.
5795. The method of item 5749 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
5796. The method of item 5749 wherein the agent is an IL-2 inhibitor.
5797. The method of item 5749 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5798. The method of item 5749 wherein the agent is an IMPDH (inosine monophosphate).
5799. The method of item 5749 wherein the agent is an integrin antagonist.
5800. The method of item 5749 wherein the agent is an interleukin antagonist.
5801. The method of item 5749 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
5802. The method of item 5749 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5803. The method of item 5749 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
5804. The method of item 5749 wherein the agent a JAK3 enzyme inhibitor.
5805. The method of item 5749 wherein the agent is a JNK inhibitor.
5806. The method of item 5749 wherein the agent is a kinase inhibitor. 5807. The method of item 5749 wherein the agent is kinesin antagonist.
5808. The method of item 5749 wherein the agent is a kinesin antagonist.
5809. The method of item 5749 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
5810. The method of item 5749 wherein the agent is an MAP kinase inhibitor.
5811. The method of item 5749 wherein the agent is a matrix metalloproteinase inhibitor.
5812. The method of item 5749 wherein the agent is an MCP-
CCR2 inhibitor.
5813. The method of item 5749 wherein the agent is an mTOR inhibitor. 5814. The method of item 5749 wherein the agent is an mTOR kinase inhibitor.
5815. The method of item 5749 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
5816. The method of item 5749 wherein the agent is an MIF inhibitor.
5817. The method of item 5749 wherein the agent is an MMP inhibitor.
5818. The method of item 5749 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 5819. The method of item 5749 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
5820. The method of item 5749 wherein the agent is a nitric oxide agonist.
5821. The method of item 5749 wherein the agent is an ornithine decarboxylase inhibitor.
5822. The method of item 5749 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5823. The method of item 5749 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
5824. The method of item 5749 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
5825. The method of item 5749 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglϊtazar (AstraZeneca), and an analogue or derivative thereof.
5826. The method of item 5749 wherein the agent is a phosphatase inhibitor.
5827. The method of item 5749 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5828. The method of item 5749 wherein the agent is a PKC inhibitor.
5829. The method of item 5749 wherein the agent is a platelet activating factor antagonist.
5830. The method of item 5749 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
5831. The method of item 5749 wherein the agent is a prolyl hydroxylase inhibitor.
5832. The method of item 5749 wherein the agent is a polymorphonuclear neutrophil inhibitor.
5833. The method of item 5749 wherein the agent is a protein kinase B inhibitor. 5834. The method of item 5749 wherein the agent is a protein kinase C stimulant.
5835. The method of item 5749 wherein the agent is a purine nucleoside analogue.
5836. The method of item 5749 wherein the agent is a purinoreceptor P2X antagonist.
5837. The method of item 5749 wherein the agent is a Raf kinase inhibitor.
5838. The method of item 5749 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
5839. The method of item 5749 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5840. The method of item 5749 wherein the agent is an SDF-1 antagonist.
5841. The method of item 5749 wherein the agent is a sheddase inhibitor.
5842. The method of item 5749 wherein the agent is an SRC inhibitor.
5843. The method of item 5749 wherein the agent is a stromelysin inhibitor.
5844. The method of item 5749 wherein the agent is an Syk kinase inhibitor.
5845. The method of item 5749 wherein the agent is a telomerase inhibitor. 5846. The method of item 5749 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5847. The method of item 5749 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGlX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB)1 apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5848. The method of item 5749 wherein the agent is a Toll receptor inhibitor.
5849. The method of item 5749 wherein the agent is a tubulin antagonist.
5850. The method of item 5749 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 5851. The method of item 5749 wherein the agent is a VEGF inhibitor.
5852. The method of item 5749 wherein the agent is a vitamin D receptor agonist.
5853. The method of item 5749 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
5854. The method of item 5749 wherein the agent is AP-23573 (an mTOR inhibitor).
5855. The method of item 5749 wherein the agent is synthadotin (a tubulin antagonist).
5856. The method of item 5749 wherein the agent is S-0885 (a collagenase inhibitor).
5857. The method of item 5749 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
5858. The method of item 5749 wherein the agent is ixabepilone (an epithilone).
5859. The method of item 5749 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
5860. The method of item 5749 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
5861. The method of item 5749 wherein the agent is ABT-518 (an angiogenesis inhibitor).
5862. The method of item 5749 wherein the agent is combretastatin (an angiogenesis inhibitor). 5863. The method of item 5749 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
5864. The method of item 5749 wherein the agent is SB- 715992 (a kinesin antagonist).
5865. The method of item 5749 wherein the agent is temsirolimus (an mTOR inhibitor).
5866. The method of item 5749 wherein the agent is adalimumab (a TNFα antagonist).
5867. The method of item 5749, wherein the composition comprises a polymer.
5868. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
5869. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
5870. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
5871. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
5872. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
5873. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer. 5874. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
5875. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
5876. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
5877. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
5878. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
5879. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
5880. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
5881. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
5882. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
5883. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer. 5884. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
5885. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
5886. The method of item 5749, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
5887. The method of item 5749, wherein the composition further comprises a second pharmaceutically active agent.
5888. The method of item 5749, wherein the composition further comprises an anti-inflammatory agent.
5889. The method of item 5749, wherein the composition further comprises an agent that inhibits infection.
5890. The method of item 5749, wherein the composition further comprises an anthracycline.
5891. The method of item 5749, wherein the composition further comprises doxorubicin.
5892. The method of item 5749 wherein the composition further comprises mitoxantrone.
5893. The method of item 5749 wherein the composition further comprises a fluoropyrimidine.
5894. The method of item 5749, wherein the composition further comprises 5-fluorouracil (5-FU).
5895. The method of item 5749, wherein the composition further comprises a folic acid antagonist. 5896. The method of item 5749, wherein the composition further comprises methotrexate.
5897. The method of item 5749, wherein the composition further comprises a podophylotoxin.
5898. The method of item 5749, wherein the composition further comprises etoposide.
5899. The method of item 5749, wherein the composition further comprises camptothecin.
5900. The method of item 5749, wherein the composition further comprises a hydroxyurea.
5901. The method of item 5749, wherein the composition further comprises a platinum complex.
5902. The method of item 5749, wherein the composition further comprises cisplatin.
5903. The method of item 5749 wherein the composition further comprises an antithrombotic agent.
5904. The method of item 5749, wherein the composition further comprises a visualization agent.
5905. The method of item 5749, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
5906. The method of item 5749, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium. further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
5908. The method of item 5749, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
5909. The method of item 5749, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
5910. The method of item 5749, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
5911. The method of item 5749, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
5912. The method of item 5749 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
5913. The method of item 5749 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
5914. The method of item 5749 wherein the composition further comprises an inflammatory cytokine.
5915. The method of item 5749 wherein the composition further comprises an agent that stimulates cell proliferation. 5916. The method of item 5749 wherein the composition further comprises a polymeric carrier.
5917. The method of item 5749 wherein the composition is in the form of a gel, paste, or spray.
5918. The method of item 5749 wherein the sensor is partially constructed with the agent or the composition.
5919. The method of item 5749 wherein the sensor is impregnated with the agent or the composition.
5920. The method of item 5749, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
5921. The method of item 5749, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
5922. The method of item 5749 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
5923. The method of item 5749, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
5924. The method of item 5749 wherein the agent or the composition is located within pores or holes of the sensor.
5925. The method of item 5749 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
5926. The method of item 5749 wherein the sensor further comprises an echogenic material.
5927. The method of item 5749 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating. 5928. The method of item 5749 wherein the sensor is sterile.
5929. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
5930. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
5931. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
5932. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
5933. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
5934. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
5935. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
5936. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days. 5937. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
5938. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
5939. The method of item 5749 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
5940. The method of item 5749 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
5941. The method of item 5749 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
5942. The method of item 5749 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
5943. The method of item 5749 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
5944. The method of item 5749 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
5945. The method of item 5749 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied. 5946. The method of item 5749 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
5947. The method of item 5749 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
5948. The method of item 5749 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
5949. The method of item 5749 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
5950. The method of item 5749 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
5951. The method of item 5749, wherein the sensor further comprises a coating, and the coating is a uniform coating.
5952. The method of item 5749, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
5953. The method of item 5749, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
5954. The method of item 5749, wherein the sensor further comprises a coating, and the coating is a patterned coating. 5955. The method of item 5749, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
5956. The method of item 5749, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
5957. The method of item 5749, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
5958. The method of item 5749, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
5959. The method of item 5749, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
5960. The method of item 5749, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5961. The method of item 5749, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5962. The method of item 5749, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
5963. The method of item 5749, wherein the sensor further comprises a coating, and the coating comprises a polymer. 5964. The method of item 5749, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
5965. The method of item 5749, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
5966. The method of item 5749 wherein the agent or the composition is affixed to the sensor.
5967. The method of item 5749 wherein the agent or the composition is covalently attached to the sensor.
5968. The method of item 5749 wherein the agent or the composition is non-covalently attached to the sensor.
5969. The method of item 5749 wherein the sensor comprises a coating that absorbs the agent or the composition.
5970. The method of item 5749 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
5971. The method of item 5749 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
5972. The method of item 5749 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
5973. The method of item 5749 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
5974. The method of item 5749 wherein the sensor is completely covered with a mesh that contains the agent or the composition. 5975. The method of item 5749 wherein the sensor is linked to a pump.
5976. The method of item 5749 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
5977. The method of item 5749 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
5978. The method of item 5749 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
5979. The method of item 5749 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
5980. The method of item 5749 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
5981. The method of item 5749 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
5982. The method of item 5749 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
5983. The method of item 5749 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor. 5984. The method of any one of items 5749-5983 wherein the pump is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site.
5985. A method for inhibiting scarring comprising placing a drug delivery implant (i.e., a pump) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5986. The method of item 5985 wherein the agent is an adensosine A2A receptor antagonist.
5987. The method of item 5985 wherein the agent is an AKT inhibitor.
5988. The method of item 5985 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5989. The method of item 5985 wherein the agent is an alpha 4 integrin antagonist.
5990. The method of item 5985 wherein the agent is an alpha 7 nicotinic receptor agonist.
5991. The method of item 5985 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKIine), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKIine), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
5992. The method of item 5985 wherein the agent is an apoptosis antagonist.
5993. The method of item 5985 wherein the agent is an apoptosis activator.
5994. The method of item 5985 wherein the agent is a beta 1 integrin antagonist.
5995. The method of item 5985 wherein the agent is a beta tubulin inhibitor. 5996. The method of item 5985 wherein the agent is a blocker of enzyme production in Hepatitis C.
5997. The method of item 5985 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5998. The method of item 5985 wherein the agent is a calcineurin inhibitor.
5999. The method of item 5985 wherein the agent is a caspase 3 inhibitor.
6000. The method of item 5985 wherein the agent is a CC chemokine receptor antagonist.
6001. The method of item 5985 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6002. The method of item 5985 wherein the agent is a cathepsin B inhibitor.
6003. The method of item 5985 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
6004. The method of item 5985 wherein the agent is a cathepsin L inhibitor.
6005. The method of item 5985 wherein the agent is a CD40 antagonist.
6006. The method of item 5985 wherein the agent is a chemokine receptor agonist. 6007. The method of item 5985 wherein the agent is a chymase inhibitor.
6008. The method of item 5985 wherein the agent is a collagenase antagonist.
6009. The method of item 5985 wherein the agent is a CXCR antagonist.
6010. The method of item 5985 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
6011. The method of item 5985 wherein the agent is a cyclooxygenase 1 inhibitor.
6012. The method of item 5985 wherein the agent is a DHFR inhibitor.
6013. The method of item 5985 wherein the agent is a dual integrin inhibitor.
6014. The method of item 5985 wherein the agent is an elastase inhibitor.
6015. The method of item 5985 wherein the agent is an elongation factor-1 alpha inhibitor. 6016. The method of item 5985 wherein the agent is an endothelial growth factor antagonist.
6017. The method of item 5985 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
6018. The method of item 5985 wherein the agent is an endotoxin antagonist.
6019. The method of item 5985 wherein the agent is an epothilone and tubulin binder.
6020. The method of item 5985 wherein the agent is an estrogen receptor antagonist.
6021. The method of item 5985 wherein the agent is an FGF inhibitor.
6022. The method of item 5985 wherein the agent is a farnexyl transferase inhibitor.
6023. The method of item 5985 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6024. The method of item 5985 wherein the agent is an FLT-3 kinase inhibitor.
6025. The method of item 5985 wherein the agent is an FGF receptor kinase inhibitor.
6026. The method of item 5985 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6027. The method of item 5985 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
6028. The method of item 5985 wherein the agent is a histone deacetylase inhibitor.
6029. The method of item 5985 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 6030. The method of item 5985 wherein the agent is an ICAM inhibitor.
6031. The method of item 5985 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6032. The method of item 5985 wherein the agent is an IL-2 inhibitor.
6033. The method of item 5985 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6034. The method of item 5985 wherein the agent is an IMPDH (inosine monophosphate). 6035. The method of item 5985 wherein the agent is an integrin antagonist.
6036. The method of item 5985 wherein the agent is an interleukin antagonist.
6037. The method of item 5985 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
6038. The method of item 5985 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
6039. The method of item 5985 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6040. The method of item 5985 wherein the agent a JAK3 enzyme inhibitor.
6041. The method of item 5985 wherein the agent is a JNK inhibitor.
6042. The method of item 5985 wherein the agent is a kinase inhibitor.
6043. The method of item 5985 wherein the agent is kinesin antagonist.
6044. The method of item 5985 wherein the agent is a kinesin antagonist.
6045. The method of item 5985 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No.D 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6046. The method of item 5985 wherein the agent is an MAP kinase inhibitor.
6047. The method of item 5985 wherein the agent is a matrix metalloproteinase inhibitor.
6048. The method of item 5985 wherein the agent is an MCP-
CCR2 inhibitor.
6049. The method of item 5985 wherein the agent is an mTOR inhibitor.
6050. The method of item 5985 wherein the agent is an mTOR kinase inhibitor.
6051. The method of item 5985 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6052. The method of item 5985 wherein the agent is an MIF inhibitor.
6053. The method of item 5985 wherein the agent is an MMP inhibitor.
6054. The method of item 5985 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6055. The method of item 5985 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 6056. The method of item 5985 wherein the agent is a nitric oxide agonist.
6057. The method of item 5985 wherein the agent is an ornithine decarboxylase inhibitor.
6058. The method of item 5985 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6059. The method of item 5985 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6060. The method of item 5985 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
6061. The method of item 5985 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6062. The method of item 5985 wherein the agent is a phosphatase inhibitor.
6063. The method of item 5985 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
6064. The method of item 5985 wherein the agent is a PKC inhibitor.
6065. The method of item 5985 wherein the agent is a platelet activating factor antagonist.
6066. The method of item 5985 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
6067. The method of item 5985 wherein the agent is a prolyl hydroxylase inhibitor.
6068. The method of item 5985 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6069. The method of item 5985 wherein the agent is a protein kinase B inhibitor.
6070. The method of item 5985 wherein the agent is a protein kinase C stimulant.
6071. The method of item 5985 wherein the agent is a purine nucleoside analogue.
6072. The method of item 5985 wherein the agent is a purinoreceptor P2X antagonist.
6073. The method of item 5985 wherein the agent is a Raf kinase inhibitor. 6074. The method of item 5985 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
6075. The method of item 5985 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
6076. The method of item 5985 wherein the agent is an SDF-1 antagonist.
6077. The method of item 5985 wherein the agent is a sheddase inhibitor.
6078. The method of item 5985 wherein the agent is an SRC inhibitor.
6079. The method of item 5985 wherein the agent is a stromelysin inhibitor.
6080. The method of item 5985 wherein the agent is an Syk kinase inhibitor.
6081. The method of item 5985 wherein the agent is a telomerase inhibitor.
6082. The method of item 5985 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
6083. The method of item 5985 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISlS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
6084. The method of item 5985 wherein the agent is a Toll receptor inhibitor.
6085. The method of item 5985 wherein the agent is a tubulin antagonist.
6086. The method of item 5985 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6087. The method of item 5985 wherein the agent is a VEGF inhibitor.
6088. The method of item 5985 wherein the agent is a vitamin D receptor agonist.
6089. The method of item 5985 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
6090. The method of item 5985 wherein the agent is AP-23573 (an mTOR inhibitor). 6091. The method of item 5985 wherein the agent is synthadotin (a tubulin antagonist).
6092. The method of item 5985 wherein the agent is S-0885 (a collagenase inhibitor).
6093. The method of item 5985 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
6094. The method of item 5985 wherein the agent is ixabepilone (an epithilone).
6095. The method of item 5985 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
6096. The method of item 5985 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6097. The method of item 5985 wherein the agent is ABT-518 (an angiogenesis inhibitor).
6098. The method of item 5985 wherein the agent is combretastatin (an angiogenesis inhibitor).
6099. The method of item 5985 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6100. The method of item 5985 wherein the agent is SB- 715992 (a kinesin antagonist).
6101. The method of item 5985 wherein the agent is temsirolimus (an mTOR inhibitor).
6102. The method of item 5985 wherein the agent is adalimumab (a TNFα antagonist). 6103. The method of item 5985, wherein the composition comprises a polymer.
6104. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
6105. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
6106. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
6107. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
6108. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
6109. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
6110. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
6111. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
6112. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 6113. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
6114. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
6115. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
6116. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
6117. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
6118. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
6119. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
6120. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
6121. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
6122. The method of item 5985, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
6123. The method of item 5985, wherein the composition further comprises a second pharmaceutically active agent. 6124. The method of item 5985, wherein the composition further comprises an anti-inflammatory agent.
6125. The method of item 5985, wherein the composition further comprises an agent that inhibits infection.
6126. The method of item 5985, wherein the composition further comprises an anthracycline.
6127. The method of item 5985, wherein the composition further comprises doxorubicin.
6128. The method of item 5985 wherein the composition further comprises mitoxantrone.
6129. The method of item 5985 wherein the composition further comprises a fluoropyrimidine.
6130. The method of item 5985, wherein the composition further comprises 5-fluorouracil (5-FU).
6131. The method of item 5985, wherein the composition further comprises a folic acid antagonist.
6132. The method of item 5985, wherein the composition further comprises methotrexate.
6133. The method of item 5985, wherein the composition further comprises a podophylotoxin.
6134. The method of item 5985, wherein the composition further comprises etoposide.
6135. The method of item 5985, wherein the composition further comprises camptothecin. 6136. The method of item 5985, wherein the composition further comprises a hydroxyurea.
6137. The method of item 5985, wherein the composition further comprises a platinum complex.
6138. The method of item 5985, wherein the composition further comprises cisplatin.
6139. The method of item 5985 wherein the composition further comprises an anti-thrombotic agent.
6140. The method of item 5985, wherein the composition further comprises a visualization agent.
6141. The method of item 5985, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
6142. The method of item 5985, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
6143. The method of item 5985, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
6144. The method of item 5985, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
6145. The method of item 5985, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 6146. The method of item 5985, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
6147. The method of item 5985, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
6148. The method of item 5985 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
6149. The method of item 5985 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
6150. The method of item 5985 wherein the composition further comprises an inflammatory cytokine.
6151. The method of item 5985 wherein the composition further comprises an agent that stimulates cell proliferation.
6152. The method of item 5985 wherein the composition further comprises a polymeric carrier.
6153. The method of item 5985 wherein the composition is in the form of a gel, paste, or spray.
6154. The method of item 5985 wherein the sensor is partially constructed with the agent or the composition.
6155. The method of item 5985 wherein the sensor is impregnated with the agent or the composition. 6156. The method of item 5985, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
6157. The method of item 5985, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
6158. The method of item 5985 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
6159. The method of item 5985, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
6160. The method of item 5985 wherein the agent or the composition is located within pores or holes of the sensor.
6161. The method of item 5985 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
6162. The method of item 5985 wherein the sensor further comprises an echogenic material.
6163. The method of item 5985 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
6164. The method of item 5985 wherein the sensor is sterile.
6165. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
6166. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue. 6167. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
6168. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
6169. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
6170. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year.
6171. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
6172. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
6173. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
6174. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate. 6175. The method of item 5985 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
6176. The method of item 5985 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
6177. The method of item 5985 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
6178. The method of item 5985 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent.
6179. The method of item 5985 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
6180. The method of item 5985 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
6181. The method of item 5985 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
6182. The method of item 5985 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied. 6183. The method of item 5985 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
6184. The method of item 5985 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
6185. The method of item 5985 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied.
6186. The method of item 5985 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
6187. The method of item 5985, wherein the sensor further comprises a coating, and the coating is a uniform coating.
6188. The method of item 5985, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
6189. The method of item 5985, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
6190. The method of item 5985, wherein the sensor further comprises a coating, and the coating is a patterned coating.
6191. The method of item 5985, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less. 6192. The method of item 5985, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
6193. The method of item 5985, wherein the sensor further comprises a coating, and the coating adheres, to the surface of the sensor upon deployment of the sensor.
6194. The method of item 5985, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
6195. The method of item 5985, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
6196. The method of item 5985, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
6197. The method of item 5985, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
6198. The method of item 5985, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
6199. The method of item 5985, wherein the sensor further comprises a coating, and the coating comprises a polymer.
6200. The method of item 5985, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition. 6201. The method of item 5985, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
6202. The method of item 5985 wherein the agent or the composition is affixed to the sensor.
6203. The method of item 5985 wherein the agent or the composition is covalently attached to the sensor.
6204. The method of item 5985 wherein the agent or the composition is non-covalently attached to the sensor.
6205. The method of item 5985 wherein the sensor comprises a coating that absorbs the agent or the composition.
6206. The method of item 5985 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
6207. The method of item 5985 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
6208. The method of item 5985 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
6209. The method of item 5985 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
6210. The method of item 5985 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
6211. The method of item 5985 wherein the sensor is linked to a pump. 6212. The method of item 5985 wherein the agent or the composition is applied to the sensor surface prior to the placing of the sensor into the host.
6213. The method of item 5985 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
6214. The method of item 5985 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
6215. The method of item 5985 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
6216. The method of item 5985 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
6217. The method of item 5985 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
6218. The method of item 5985 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
6219. The method of item 5985 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
6220. A method for making a device comprising: combining a sensor and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
6221. The method of item 6220 wherein the agent is an adensosine A2A receptor antagonist.
6222. The method of item 6220 wherein the agent is an AKT inhibitor.
6223. The method of item 6220 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
6224. The method of item 6220 wherein the agent is an alpha 4 integrin antagonist.
6225. The method of item 6220 wherein the agent is an alpha 7 nicotinic receptor agonist.
6226. The method of item 6220 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6227. The method of item 6220 wherein the agent is an apoptosis antagonist.
6228. The method of item 6220 wherein the agent is an apoptosis activator.
6229. The method of item 6220 wherein the agent is a beta 1 integrin antagonist.
6230. The method of item 6220 wherein the agent is a beta tubulin inhibitor.
6231. The method of item 6220 wherein the agent is a blocker of enzyme production in Hepatitis C.
6232. The method of item 6220 wherein the agent is a Bruton's tyrosine kinase inhibitor. 6233. The method of item 6220 wherein the agent is a calcineurin inhibitor.
6234. The method of item 6220 wherein the agent is a caspase 3 inhibitor.
6235. The method of item 6220 wherein the agent is a CC chemokine receptor antagonist.
6236. The method of item 6220 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6237. The method of item 6220 wherein the agent is a cathepsin B inhibitor.
6238. The method of item 6220 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
6239. The method of item 6220 wherein the agent is a cathepsin L inhibitor.
6240. The method of item 6220 wherein the agent is a CD40 antagonist.
6241. The method of item 6220 wherein the agent is a chemokine receptor agonist.
6242. The method of item 6220 wherein the agent is a chymase inhibitor.
6243. The method of item 6220 wherein the agent is a collagenase antagonist. 6244. The method of item 6220 wherein the agent is a CXCR antagonist.
6245. The method of item 6220 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
6246. The method of item 6220 wherein the agent is a cyclooxygenase 1 inhibitor.
6247. The method of item 6220 wherein the agent is a DHFR inhibitor.
6248. The method of item 6220 wherein the agent is a dual integrin inhibitor.
6249. The method of item 6220 wherein the agent is an elastase inhibitor.
6250. The method of item 6220 wherein the agent is an elongation factor-1 alpha inhibitor.
6251. The method of item 6220 wherein the agent is an endothelial growth factor antagonist.
6252. The method of item 6220 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
6253. The method of item 6220 wherein the agent is an endotoxin antagonist.
6254. The method of item 6220 wherein the agent is an epothilone and tubulin binder.
6255. The method of item 6220 wherein the agent is an estrogen receptor antagonist.
6256. The method of item 6220 wherein the agent is an FGF inhibitor.
6257. The method of item 6220 wherein the agent is a famexyl transferase inhibitor.
6258. The method of item 6220 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6259. The method of item 6220 wherein the agent is an FLT-3 kinase inhibitor. 6260. The method of item 6220 wherein the agent is an FGF receptor kinase inhibitor.
6261. The method of item 6220 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6262. The method of item 6220 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005
(Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
6263. The method of item 6220 wherein the agent is a histone deacetylase inhibitor.
6264. The method of item 6220 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
6265. The method of item 6220 wherein the agent is an ICAM inhibitor.
6266. The method of item 6220 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 6267. The method of item 6220 wherein the agent is an IL-2 inhibitor.
6268. The method of item 6220 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6269. The method of item 6220 wherein the agent is an IMPDH (inosine monophosphate).
6270. The method of item 6220 wherein the agent is an integrin antagonist.
6271. The method of item 6220 wherein the agent is an interleukin antagonist.
6272. The method of item 6220 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 6273. The method of item 6220 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
6274. The method of item 6220 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6275. The method of item 6220 wherein the agent a JAK3 enzyme inhibitor.
6276. The method of item 6220 wherein the agent is a JNK inhibitor.
6277. The method of item 6220 wherein the agent is a kinase inhibitor.
6278. The method of item 6220 wherein the agent is kinesin antagonist.
6279. The method of item 6220 wherein the agent is a kinesin antagonist.
6280. The method of item 6220 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucothene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6281. The method of item 6220 wherein the agent is an MAP kinase inhibitor.
6282. The method of item 6220 wherein the agent is a matrix metalloproteinase inhibitor.
6283. The method of item 6220 wherein the agent is an MCP- CCR2 inhibitor.
6284. The method of item 6220 wherein the agent is an mTOR inhibitor.
6285. The method of item 6220 wherein the agent is an mTOR kinase inhibitor.
6286. The method of item 6220 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6287. The method of item 6220 wherein the agent is an MIF inhibitor. 6288. The method of item 6220 wherein the agent is an MMP inhibitor.
6289. The method of item 6220 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6290. The method of item 6220 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium
Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
6291. The method of item 6220 wherein the agent is a nitric oxide agonist.
6292. The method of item 6220 wherein the agent is an ornithine decarboxylase inhibitor.
6293. The method of item 6220 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p3δ-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6294. The method of item 6220 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6295. The method of item 6220 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
6296. The method of item 6220 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET
(GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6297. The method of item 6220 wherein the agent is a phosphatase inhibitor.
6298. The method of item 6220 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 6299. The method of item 6220 wherein the agent is a PKC inhibitor.
6300. The method of item 6220 wherein the agent is a platelet activating factor antagonist.
6301. The method of item 6220 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
6302. The method of item 6220 wherein the agent is a prolyl hydroxylase inhibitor.
6303. The method of item 6220 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6304. The method of item 6220 wherein the agent is a protein kinase B inhibitor.
6305. The method of item 6220 wherein the agent is a protein kinase C stimulant.
6306. The method of item 6220 wherein the agent is a purine nucleoside analogue.
6307. The method of item 6220 wherein the agent is a purinoreceptor P2X antagonist.
6308. The method of item 6220 wherein the agent is a Raf kinase inhibitor.
6309. The method of item 6220 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
6310. The method of item 6220 wherein the agent is a ribonucleoside triphosphate reductase inhibitor. 6311. The method of item 6220 wherein the agent is an SDF-1 antagonist.
6312. The method of item 6220 wherein the agent is a sheddase inhibitor.
6313. The method of item 6220 wherein the agent is an SRC inhibitor.
6314. The method of item 6220 wherein the agent is a stromelysin inhibitor.
6315. The method of item 6220 wherein the agent is an Syk kinase inhibitor.
6316. The method of item 6220 wherein the agent is a telomerase inhibitor.
6317. The method of item 6220 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
6318. The method of item 6220 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 6319. The method of item 6220 wherein the agent is a Toll receptor inhibitor.
6320. The method of item 6220 wherein the agent is a tubulin antagonist.
6321. The method of item 6220 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis
Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6322. The method of item 6220 wherein the agent is a VEGF inhibitor.
6323. The method of item 6220 wherein the agent is a vitamin D receptor agonist.
6324. The method of item 6220 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
6325. The method of item 6220 wherein the agent is AP-23573 (an mTOR inhibitor).
6326. The method of item 6220 wherein the agent is synthadotin (a tubulin antagonist).
6327. The method of item 6220 wherein the agent is S-0885 (a collagenase inhibitor). 6328. The method of item 6220 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
6329. The method of item 6220 wherein the agent is ixabepilone (an epithilone).
6330. The method of item 6220 wherein the agent is IDN-5390
(an angiogenesis inhibitor).
6331. The method of item 6220 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6332. The method of item 6220 wherein the agent is ABT-518 (an angiogenesis inhibitor).
6333. The method of item 6220 wherein the agent is combretastatin (an angiogenesis inhibitor).
6334. The method of item 6220 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6335. The method of item 6220 wherein the agent is SB-
715992 (a kinesin antagonist).
6336. The method of item 6220 wherein the agent is temsirolimus (an mTOR inhibitor).
6337. The method of item 6220 wherein the agent is adalimumab (a TNFα antagonist).
6338. The method of item 6220, wherein the composition comprises a polymer.
6339. The method of item 6220, wherein the composition comprises a polymeric carrier. 6340. The method of item 6220 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
6341. The method of item 6220 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
6342. The method of item 6220 wherein the device has a coating that comprises the anti-scarring agent.
6343. The method of item 6220, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
6344. The method of item 6220, wherein the device has a coating that comprises the agent and directly contacts the sensor.
6345. The method of item 6220, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
6346. The method of item 6220, wherein the device has a coating that comprises the agent and partially covers the sensor.
6347. The method of item 6220, wherein the device has a coating that comprises the agent and completely covers the sensor.
6348. The method of item 6220, wherein the device has a uniform coating.
6349. The method of item 6220, wherein the device has a nonuniform coating.
6350. The method of item 6220, wherein the device has a discontinuous coating.
6351. The method of item 6220, wherein the device has a patterned coating. 6352. The method of item 6220, wherein the device has a coating with a thickness of 100 μm or less.
6353. The method of item 6220, wherein the device has a coating with a thickness of 10 μm or less.
6354. The method of item 6220, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
6355. The method of item 6220, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
6356. The method of item 6220, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1% by weight.
6357. The method of item 6220, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
6358. The method of item 6220, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
6359. The method of item 6220, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
6360. The method of item 6220, wherein the device has a coating, and wherein the coating further comprises a polymer. 6361. The method of item 6220, wherein the device has a first coating having a first composition and a second coating having a second composition.
6362. The method of item 6220, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
6363. The method of item 6220, wherein the composition comprises a polymer.
6364. The method of item 6220, wherein the composition comprises a polymeric carrier.
6365. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
6366. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
6367. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
6368. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
6369. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 6370. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
6371. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
6372. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
6373. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
6374. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
6375. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
6376. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
6377. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
6378. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer. 6379. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
6380. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
6381. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
6382. The method of item 6220, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
6383. The method of item 6220 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
6384. The method of item 6220, wherein the device comprises a lubricious coating.
6385. The method of item 6220 wherein the anti-scarring agent is located within pores or holes of the device.
6386. The method of item 6220 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
6387. The method of item 6220, wherein the device comprises a second pharmaceutically active agent.
6388. The method of item 6220 wherein the device comprises an anti-inflammatory agent. 6389. The method of item 6220 wherein the device comprises an agent that inhibits infection.
6390. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
6391. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
6392. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
6393. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
6394. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
6395. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
6396. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
6397. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
6398. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
6399. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
6400. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea. 6401. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
6402. The method of item 6220 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
6403. The method of item 6220, further comprising an antithrombotic agent.
6404. The method of item 6220 wherein the device comprises a visualization agent.
6405. The method of item 6220 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
6406. The method of item 6220 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
6407. The method of item 6220 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
6408. The method of item 6220 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
6409. The method of item 6220 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 6410. The method of item 6220 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
6411. The method of item 6220 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
6412. The method of item 6220 wherein the device comprises an echogenic material.
6413. The method of item 6220 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
6414. The method of item 6220 wherein the device is sterile.
6415. The method of item 6220 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
6416. The method of item 6220 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
6417. The method of item 6220 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
6418. The method of item 6220 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 6419. The method of item 6220 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
6420. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
6421. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
6422. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
6423. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
6424. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
6425. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
6426. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
6427. The method of item 6220 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days. 6428. The method of item 6220 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
6429. The method of item 6220 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
6430. The method of item 6220 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
6431. The method of item 6220 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
6432. The method of item 6220 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
6433. The method of item 6220 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6434. The method of item 6220 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6435. The method of item 6220 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6436. The method of item 6220 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6437. The method of item 6220 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 6438. The method of item 6220 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6439. The method of item 6220 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
6440. The method of item 6220 wherein the combining is performed by spraying the agent or the component onto the sensor.
6441. The method of item 6220 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
6442. The method of item 6220 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
6443. The method of item 6220 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
6444. The method of item 6220 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
6445. The method of item 6220 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
6446. The method of item 6220 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
6447. The method of item 6220 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 6448. The method of item 6220 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
6449. The method of item 6220 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
6450. The method of item 6220 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
6451. The method of item 6220 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
6452. The method of item 6220 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
6453. The method of item 6220 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
6454. The method of item 6220 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
6455. The method of item 6220 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
6456. The method of item 6220 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition. 6457. The method of item 6220 wherein the combining is performed by impregnating the sensor with the agent or the composition.
6458. The method of item 6220 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
6459. The method of item 6220 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
6460. The method of item 6220 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
6461. The method of item 6220 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
6462. The method of item 6220 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
6463. The method of item 6220 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
6464. The method of item 6220 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
6465. The method of item 6220 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor. 6466. The method of item 6220 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
6467. The method of item 6220 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
6468. The method of item 6220 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
6469. The method of item 6220 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
6470. The method of item 6220 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
6471. The method of any one of items 6220-6470 wherein the sensor is a blood or tissue glucose monitor.
6472. The method of any one of items 6220-6470 wherein the sensor is an electrolyte sensor.
6473. The method of any one of items 6220-6470 wherein the sensor is a blood constituent sensor.
6474. The method of any one of items 6220-6470 wherein the sensor is a temperature sensor.
6475. The method of any one of items 6220-6470 wherein the sensor is a pH sensor. 16476. The method of any one of items 6220-6470 wherein the sensor is an optical sensor.
6477. The method of any one of items 6220-6470 wherein the sensor is an amperometric sensor.
6478. The method of any one of items 6220-6470 wherein the sensor is a pressure sensor.
6479. The method of any one of items 6220-6470 wherein the sensor is a biosensor.
6480. The method of any one of items 6220-6470 wherein the sensor is a sensing transponder.
)
6481. The method of any one of items 6220-6470 wherein the sensor is a strain sensor.
6482. The method of any one of items 6220-6470 wherein the sensor is a magnetoresistive sensor.
6483. The method of any one of items 6220-6470 wherein the sensor is a cardiac sensor.
6484. The method of any one of items 6220-6470 wherein the sensor is a respiratory sensor.
6485. The method of any one of items 6220-6470 wherein the sensor is an auditory sensor.
6486. The method of any one of items 6220-6470 wherein the sensor is a metabolite sensor.
6487. The method of any one of items 6220-6470 wherein the sensor detects mechanical changes. 6488. The method of any one of items 6220-6470 wherein the sensor detects physical changes.
6489. The method of any one of items 6220-6470 wherein the sensor detects electrochemical changes.
6490. The method of any one of items 6220-6470 wherein the sensor detects magnetic changes.
6491. The method of any one of items 6220-6470 wherein the sensor detects acceleration changes.
6492. The method of any one of items 6220-6470 wherein the sensor detects ionizing radiation changes.
6493. The method of any one of items 6220-6470 wherein the sensor detects acoustic wave changes.
6494(. The method of any one of items 6220-6470 wherein the sensor detects chemical changes.
6495. The method of any one of items 6220-6470 wherein the sensor detects drug concentration changes.
6496. The method of any one of items 6220-6470 wherein the sensor detects hormone changes.
6497. The method of any one of items 6220-6470 wherein the sensor detects barometric changes.
6498. A method for making a device comprising: combining a blood or tissue glucose monitor (i.e., a sensor) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 6499. The method of item 6498 wherein the agent is an adensosine A2A receptor antagonist.
6500. The method of item 6498 wherein the agent is an AKT inhibitor.
6501. The method of item 6498 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
6502. The method of item 6498 wherein the agent is an alpha 4 integrin antagonist.
6503. The method of item 6498 wherein the agent is an alpha 7 nicotinic receptor agonist.
6504. The method of item 6498 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant
Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-
23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6505. The method of item 6498 wherein the agent is an apoptosis antagonist.
6506. The method of item 6498 wherein the agent is an apoptosis activator.
6507. The method of item 6498 wherein the agent is a beta 1 integrin antagonist.
6508. The method of item 6498 wherein the agent is a beta tubulin inhibitor.
6509. The method of item 6498 wherein the agent is a blocker of enzyme production in Hepatitis C.
6510. The method of item 6498 wherein the agent is a Bruton's tyrosine kinase inhibitor.
6511. The method of item 6498 wherein the agent is a calcineurin inhibitor. 6512. The method of item 6498 wherein the agent is a caspase 3 inhibitor.
6513. The method of item 6498 wherein the agent is a CC chemokine receptor antagonist.
6514. The method of item 6498 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6515. The method of item 6498 wherein the agent is a cathepsin B inhibitor.
6516. The method of item 6498 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GiaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
6517. The method of item 6498 wherein the agent is a cathepsin L inhibitor.
6518. The method of item 6498 wherein the agent is a CD40 antagonist.
6519. The method of item 6498 wherein the agent is a chemokine receptor agonist.
6520. The method of item 6498 wherein the agent is a chymase inhibitor.
6521. The method of item 6498 wherein the agent is a collagenase antagonist.
6522. The method of item 6498 wherein the agent is a CXCR antagonist. 6523. The method of item 6498 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
6524. The method of item 6498 wherein the agent is a cyclooxygenase 1 inhibitor.
6525. The method of item 6498 wherein the agent is a DHFR inhibitor.
6526. The method of item 6498 wherein the agent is a dual integrin inhibitor.
6527. The method of item 6498 wherein the agent is an elastase inhibitor.
6528. The method of item 6498 wherein the agent is an elongation factor-1 alpha inhibitor.
6529. The method of item 6498 wherein the agent is an endothelial growth factor antagonist.
6530. The method of item 6498 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
6531. The method of item 6498 wherein the agent is an endotoxin antagonist.
6532. The method of item 6498 wherein the agent is an epothilone and tubulin binder.
6533. The method of item 6498 wherein the agent is an estrogen receptor antagonist.
6534. The method of item 6498 wherein the agent is an FGF inhibitor.
6535. The method of item 6498 wherein the agent is a famexyl transferase inhibitor.
6536. The method of item 6498 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6537. The method of item 6498 wherein the agent is an FLT-3 kinase inhibitor.
6538. The method of item 6498 wherein the agent is an FGF receptor kinase inhibitor. 6539. The method of item 6498 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6540. The method of item 6498 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethyIaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1I,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
6541. The method of item 6498 wherein the agent is a histone deacetylase inhibitor.
6542. The method of item 6498 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
6543. The method of item 6498 wherein the agent is an ICAM inhibitor.
6544. The method of item 6498 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6545. The method of item 6498 wherein the agent is an IL-2 inhibitor. 6546. The method of item 6498 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6547. The method of item 6498 wherein the agent is an IMPDH (inosine monophosphate).
6548. The method of item 6498 wherein the agent is an integrin antagonist.
6549. The method of item 6498 wherein the agent is an interleukin antagonist.
6550. The method of item 6498 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
6551. The method of item 6498 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 6552. The method of item 6498 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6553. The method of item 6498 wherein the agent a JAK3 enzyme inhibitor.
6554. The method of item 6498 wherein the agent is a JNK inhibitor.
6555. The method of item 6498 wherein the agent is a kinase inhibitor.
6556. The method of item 6498 wherein the agent is kinesin antagonist.
6557. The method of item 6498 wherein the agent is a kinesin antagonist.
6558. The method of item 6498 wherein the agent is a leukothene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No.
346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof. 6559. The method of item 6498 wherein the agent is an MAP kinase inhibitor.
6560. The method of item 6498 wherein the agent is a matrix metalloproteinase inhibitor.
6561. The method of item 6498 wherein the agent is an MCP-
CCR2 inhibitor.
6562. The method of item 6498 wherein the agent is an mTOR inhibitor.
6563. The method of item 6498 wherein the agent is an mTOR kinase inhibitor.
6564. The method of item 6498 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6565. The method of item 6498 wherein the agent is an MIF inhibitor.
6566. The method of item 6498 wherein the agent is an MMP inhibitor. 6567. The method of item 6498 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6568. The method of item 6498 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-
576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
6569. The method of item 6498 wherein the agent is a nitric oxide agonist.
6570. The method of item 6498 wherein the agent is an ornithine decarboxylase inhibitor.
6571. The method of item 6498 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6572. The method of item 6498 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6573. The method of item 6498 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
6574. The method of item 6498 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6575. The method of item 6498 wherein the agent is a phosphatase inhibitor.
6576. The method of item 6498 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
6577. The method of item 6498 wherein the agent is a PKC inhibitor.
6578. The method of item 6498 wherein the agent is a platelet activating factor antagonist. 6579. The method of item 6498 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
6580. The method of item 6498 wherein the agent is a prolyl hydroxylase inhibitor.
6581. The method of item 6498 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6582. The method of item 6498 wherein the agent is a protein kinase B inhibitor.
6583. The method of item 6498 wherein the agent is a protein kinase C stimulant.
6584. The method of item 6498 wherein the agent is a purine nucleoside analogue.
6585. The method of item 6498 wherein the agent is a purinoreceptor P2X antagonist.
6586. The method of item 6498 wherein the agent is a Raf kinase inhibitor.
6587. The method of item 6498 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
6588. The method of item 6498 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
6589. The method of item 6498 wherein the agent is an SDF-1 antagonist.
6590. The method of item 6498 wherein the agent is a sheddase inhibitor. 6591. The method of item 6498 wherein the agent is an SRC inhibitor.
6592. The method of item 6498 wherein the agent is a stromelysin inhibitor.
6593. The method of item 6498 wherein the agent is an Syk kinase inhibitor.
6594. The method of item 6498 wherein the agent is a telomerase inhibitor.
6595. The method of item 6498 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No.
53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
6596. The method of item 6498 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
6597. The method of item 6498 wherein the agent is a Toll receptor inhibitor.
6598. The method of item 6498 wherein the agent is a tubulin antagonist. 6599. The method of item 6498 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6600. The method of item 6498 wherein the agent is a VEGF inhibitor.
6601. The method of item 6498 wherein the agent is a vitamin D receptor agonist.
6602. The method of item 6498 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
6603. The method of item 6498 wherein the agent is AP-23573 (an mTOR inhibitor).
6604. The method of item 6498 wherein the agent is synthadotin (a tubulin antagonist).
6605. The method of item 6498 wherein the agent is S-0885 (a collagenase inhibitor).
6606. The method of item 6498 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
6607. The method of item 6498 wherein the agent is ixabepilone (an epithilone). 6608. The method of item 6498 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
6609. The method of item 6498 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6610. The method of item 6498 wherein the agent is ABT-518
(an angiogenesis inhibitor).
6611. The method of item 6498 wherein the agent is combretastatin (an angiogenesis inhibitor).
6612. The method of item 6498 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6613. The method of item 6498 wherein the agent is SB- 715992 (a kinesin antagonist).
6614. The method of item 6498 wherein the agent is temsirolimus (an mTOR inhibitor).
6615. The method of item 6498 wherein the agent is adalimumab (a TNFα antagonist).
6616. The method of item 6498, wherein the composition comprises a polymer.
6617. The method of item 6498, wherein the composition comprises a polymeric carrier.
6618. The method of item 6498 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
6619. The method of item 6498 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 6620. The method of item 6498 wherein the device has a coating that comprises the anti-scarring agent.
6621. The method of item 6498, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
6622. The method of item 6498, wherein the device has a coating that comprises the agent and directly contacts the sensor.
6623. The method of item 6498, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
6624. The method of item 6498, wherein the device has a coating that comprises the agent and partially covers the sensor.
6625. The method of item 6498, wherein the device has a coating that comprises the agent and completely covers the sensor.
6626. The method of item 6498, wherein the device has a uniform coating.
6627. The method of item 6498, wherein the device has a nonuniform coating.
6628. The method of item 6498, wherein the device has a discontinuous coating.
6629. The method of item 6498, wherein the device has a patterned coating.
6630. The method of item 6498, wherein the device has a coating with a thickness of 100 μm or less.
6631. The method of item 6498, wherein the device has a coating with a thickness of 10 μm or less. 6632. The method of item 6498, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
6633. The method of item 6498, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
6634. The method of item 6498, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1% by weight.
6635. The method of item 6498, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
6636. The method of item 6498, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
6637. The method of item 6498, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
6638. The method of item 6498, wherein the device has a coating, and wherein the coating further comprises a polymer.
6639. The method of item 6498, wherein the device has a first coating having a first composition and a second coating having a second composition.
6640. The method of item 6498, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different. 6641. The method of item 6498, wherein the composition comprises a polymer.
6642. The method of item 6498, wherein the composition comprises a polymeric carrier.
6643. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
6644. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
6645. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
6646. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
6647. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
6648. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
6649. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer. 6650. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
6651. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
6652. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
6653. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
6654. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
6655. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
6656. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
6657. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
6658. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer. 6659. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
6660. The method of item 6498, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
6661. The method of item 6498 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
6662. The method of item 6498, wherein the device comprises a lubricious coating.
6663. The method of item 6498 wherein the anti-scarring agent is located within pores or holes of the device.
6664. The method of item 6498 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
6665. The method of item 6498, wherein the device comprises a second pharmaceutically active agent.
6666. The method of item 6498 wherein the device comprises an anti-inflammatory agent.
6667. The method of item 6498 wherein the device comprises an agent that inhibits infection.
6668. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
6669. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin. 6670. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
6671. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
6672. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
6673. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
6674. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
6675. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
6676. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
6677. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
6678. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
6679. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
6680. The method of item 6498 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
6681. The method of item 6498, further comprising an antithrombotic agent. 6682. The method of item 6498 wherein the device comprises a visualization agent.
6683. The method of item 6498 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
6684. The method of item 6498 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
6685. The method of item 6498 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
6686. The method of item 6498 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
6687. The method of item 6498 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
6688. The method of item 6498 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
6689. The method of item 6498 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
6690. The method of item 6498 wherein the device comprises an echogenic material. an ecnogenic material, ana wnerein tne ecnogenic material is in the form of a coating.
6692. The method of item 6498 wherein the device is sterile.
6693. The method of item 6498 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
6694. The method of item 6498 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
6695. The method of item 6498 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
6696. The method of item 6498 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
6697. The method of item 6498 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
6698. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
6699. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 6700. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
6701. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
6702. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
6703. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
6704. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
6705. The method of item 6498 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
6706. The method of item 6498 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
6707. The method of item 6498 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
6708. The method of item 6498 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
6709. The method of item 6498 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. 6710. The method of item 6498 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
6711. The method of item 6498 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6712. The method of item 6498 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6713. The method of item 6498 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6714. The method of item 6498 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6715. The method of item 6498 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6716. The method of item 6498 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6717. The method of item 6498 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
6718. The method of item 6498 wherein the combining is performed by spraying the agent or the component onto the sensor. 6719. The method of item 6498 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
6720. The method of item 6498 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
6721. The method of item 6498 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
6722. The method of item 6498 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
6723. The method of item 6498 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
6724. The method of item 6498 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
6725. The method of item 6498 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
6726. The method of item 6498 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
6727. The method of item 6498 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition. 6728. The method of item 6498 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
6729. The method of item 6498 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
6730. The method of item 6498 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
6731. The method of item 6498 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
6732. The method of item 6498 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
6733. The method of item 6498 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
6734. The method of item 6498 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
6735. The method of item 6498 wherein the combining is performed by impregnating the sensor with the agent or the composition.
6736. The method of item 6498 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent. 6737. The method of item 6498 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
6738. The method of item 6498 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
6739. The method of item 6498 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
6740. The method of item 6498 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
6741. The method of item 6498 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
6742. The method of item 6498 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
6743. The method of item 6498 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
6744. The method of item 6498 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
6745. The method of item 6498 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor. 6746. The method of item 6498 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
6747. The method of item 6498 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
6748. The method of item 6498 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
6749. The method of any one of items 6498-6748 wherein the device is deliverable to the vascular system transluminal^ using a catheter on a stent platform.
6750. The method of any one of items 6498-6748 wherein the device is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
6751. The method of any one of items 6498-6748 wherein the device is an electrode composed of an analyte responsive enzyme.
6752. The method of any one of items 6498-6748 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
6753. The method of any one of items 6498-6748 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin. 6754. A method for making a device comprising: combining a pressure or stress sensor and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
6755. The method of item 6754 wherein the agent is an adensosine A2A receptor antagonist.
6756. The method of item 6754 wherein the agent is an AKT inhibitor.
6757. The method of item 6754 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects
No. 5754 (Merck KgaA).
6758. The method of item 6754 wherein the agent is an alpha 4 integrin antagonist.
6759. The method of item 6754 wherein the agent is an alpha 7 nicotinic receptor agonist.
6760. The method of item 6754 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005
(GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6761. The method of item 6754 wherein the agent is an apoptosis antagonist.
6762. The method of item 6754 wherein the agent is an apoptosis activator.
6763. The method of item 6754 wherein the agent is a beta 1 integrin antagonist.
6764. The method of item 6754 wherein the agent is a beta tubulin inhibitor.
6765. The method of item 6754 wherein the agent is a blocker of enzyme production in Hepatitis C. 6766. The method of item 6754 wherein the agent is a Bruton's tyrosine kinase inhibitor.
6767. The method of item 6754 wherein the agent is a calcineurin inhibitor.
6768. The method of item 6754 wherein the agent is a caspase
3 inhibitor.
6769. The method of item 6754 wherein the agent is a CC chemokine receptor antagonist.
6770. The method of item 6754 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin,
KRX-0403, and an analogue or derivative thereof.
6771. The method of item 6754 wherein the agent is a cathepsin B inhibitor.
6772. The method of item 6754 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795
(GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
6773. The method of item 6754 wherein the agent is a cathepsin L inhibitor.
6774. The method of item 6754 wherein the agent is a CD40 antagonist.
6775. The method of item 6754 wherein the agent is a chemokine receptor agonist.
6776. The method of item 6754 wherein the agent is a chymase inhibitor. 6777. The method of item 6754 wherein the agent is a collagenase antagonist.
6778. The method of item 6754 wherein the agent is a CXCR antagonist.
6779. The method of item 6754 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
6780. The method of item 6754 wherein the agent is a cyclooxygenase 1 inhibitor.
6781. The method of item 6754 wherein the agent is a DHFR inhibitor.
6782. The method of item 6754 wherein the agent is a dual integrin inhibitor.
6783. The method of item 6754 wherein the agent is an elastase inhibitor.
6784. The method of item 6754 wherein the agent is an elongation factor-1 alpha inhibitor.
6785. The method of item 6754 wherein the agent is an endothelial growth factor antagonist. 6786. The method of item 6754 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
6787. The method of item 6754 wherein the agent is an endotoxin antagonist.
6788. The method of item 6754 wherein the agent is an epothilone and tubulin binder.
6789. The method of item 6754 wherein the agent is an estrogen receptor antagonist.
6790. The method of item 6754 wherein the agent is an FGF inhibitor.
6791. The method of item 6754 wherein the agent is a farnexyl transferase inhibitor.
6792. The method of item 6754 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme),
Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof. 6793. The method of item 6754 wherein the agent is an FLT-3 kinase inhibitor.
6794. The method of item 6754 wherein the agent is an FGF receptor kinase inhibitor.
6795. The method of item 6754 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6796. The method of item 6754 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
6797. The method of item 6754 wherein the agent is a histone deacetylase inhibitor.
6798. The method of item 6754 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
6799. The method of item 6754 wherein the agent is an ICAM inhibitor. 6800. The method of item 6754 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6801. The method of item 6754 wherein the agent is an IL-2 inhibitor.
6802. The method of item 6754 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6803. The method of item 6754 wherein the agent is an IMPDH (inosine monophosphate).
6804. The method of item 6754 wherein the agent is an integrin antagonist. 6805. The method of item 6754 wherein the agent is an interleukin antagonist.
6806. The method of item 6754 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
6807. The method of item 6754 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
6808. The method of item 6754 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6809. The method of item 6754 wherein the agent a JAK3 enzyme inhibitor.
6810. The method of item 6754 wherein the agent is a JNK inhibitor.
6811. The method of item 6754 wherein the agent is a kinase inhibitor.
6812. The method of item 6754 wherein the agent is kinesin antagonist.
6813. The method of item 6754 wherein the agent is a kinesin antagonist.
6814. The method of item 6754 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6815. The method of item 6754 wherein the agent is an MAP kinase inhibitor.
6816. The method of item 6754 wherein the agent is a matrix metalloproteinase inhibitor.
6817. The method of item 6754 wherein the agent is an MCP- CCR2 inhibitor.
6818. The method of item 6754 wherein the agent is an mTOR inhibitor.
6819. The method of item 6754 wherein the agent is an mTOR kinase inhibitor.
6820. The method of item 6754 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6821. The method of item 6754 wherein the agent is an MlF inhibitor.
6822. The method of item 6754 wherein the agent is an MMP inhibitor.
6823. The method of item 6754 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6824. The method of item 6754 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
6825. The method of item 6754 wherein the agent is a nitric oxide agonist. 6826. The method of item 6754 wherein the agent is an ornithine decarboxylase inhibitor.
6827. The method of item 6754 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6828. The method of item 6754 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6829. The method of item 6754 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
6830. The method of item 6754 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6831. The method of item 6754 wherein the agent is a phosphatase inhibitor.
6832. The method of item 6754 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
6833. The method of item 6754 wherein the agent is a PKC inhibitor.
6834. The method of item 6754 wherein the agent is a platelet activating factor antagonist.
6835. The method of item 6754 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
6836. The method of item 6754 wherein the agent is a prolyl hydroxylase inhibitor.
6837. The method of item 6754 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6838. The method of item 6754 wherein the agent is a protein kinase B inhibitor.
6839. The method of item 6754 wherein the agent is a protein kinase C stimulant.
6840. The method of item 6754 wherein the agent is a purine nucleoside analogue.
6841. The method of item 6754 wherein the agent is a purinoreceptor P2X antagonist.
6842. The method of item 6754 wherein the agent is a Raf kinase inhibitor.
6843. The method of item 6754 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 6844. The method of item 6754 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
6845. The method of item 6754 wherein the agent is an SDF-1 antagonist.
6846. The method of item 6754 wherein the agent is a sheddase inhibitor.
6847. The method of item 6754 wherein the agent is an SRC inhibitor.
6848. The method of item 6754 wherein the agent is a stromelysin inhibitor.
6849. The method of item 6754 wherein the agent is an Syk kinase inhibitor.
6850. The method of item 6754 wherein the agent is a telomerase inhibitor.
6851. The method of item 6754 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
6852. The method of item 6754 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS)1 infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
6853. The method of item 6754 wherein the agent is a Toll receptor inhibitor.
6854. The method of item 6754 wherein the agent is a tubulin antagonist.
6855. The method of item 6754 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmith Kline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6856. The method of item 6754 wherein the agent is a VEGF inhibitor.
6857. The method of item 6754 wherein the agent is a vitamin D receptor agonist.
6858. The method of item 6754 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
6859. The method of item 6754 wherein the agent is AP-23573
(an mTOR inhibitor).
6860. The method of item 6754 wherein the agent is synthadotin (a tubulin antagonist). 6861. The method of item 6754 wherein the agent is S-0885 (a collagenase inhibitor).
6862. The method of item 6754 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
6863. The method of item 6754 wherein the agent is ixabepilone (an epithilone).
6864. The method of item 6754 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
6865. The method of item 6754 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6866. The method of item 6754 wherein the agent is ABT-518 (an angiogenesis inhibitor).
6867. The method of item 6754 wherein the agent is combretastatin (an angiogenesis inhibitor).
6868. The method of item 6754 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6869. The method of item 6754 wherein the agent is SB- 715992 (a kinesin antagonist).
6870. The method of item 6754 wherein the agent is temsirolimus (an mTOR inhibitor).
6871. The method of item 6754 wherein the agent is adalimumab (a TNFα antagonist).
6872. The method of item 6754, wherein the composition comprises a polymer. 6873. The method of item 6754, wherein the composition comprises a polymeric carrier.
6874. The method of item 6754 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
6875. The method of item 6754 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
6876. The method of item 6754 wherein the device has a coating that comprises the anti-scarring agent.
6877. The method of item 6754, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
6878. The method of item 6754, wherein the device has a coating that comprises the agent and directly contacts the sensor.
6879. The method of item 6754, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
6880. The method of item 6754, wherein the device has a coating that comprises the agent and partially covers the sensor.
6881. The method of item 6754, wherein the device has a coating that comprises the agent and completely covers the sensor.
6882. The method of item 6754, wherein the device has a uniform coating.
6883. The method of item 6754, wherein the device has a nonuniform coating.
6884. The method of item 6754, wherein the device has a discontinuous coating. 6885. The method of item 6754, wherein the device has a patterned coating.
6886. The method of item 6754, wherein the device has a coating with a thickness of 100 μm or less.
6887. The method of item 6754, wherein the device has a coating with a thickness of 10 μm or less.
6888. The method of item 6754, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
6889. The method of item 6754, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
6890. The method of item 6754, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
6891. The method of item 6754, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
6892. The method of item 6754, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
6893. The method of item 6754, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
6894. The method of item 6754, wherein the device has a coating, and wherein the coating further comprises a polymer. 6895. The method of item 6754, wherein the device has a first coating having a first composition and a second coating having a second composition.
6896. The method of item 6754, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
6897. The method of item 6754, wherein the composition comprises a polymer.
6898. The method of item 6754, wherein the composition comprises a polymeric carrier.
6899. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
6900. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
6901. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
6902. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
6903. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 6904. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
6905. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
6906. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
6907. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
6908. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
6909. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
6910. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
6911. The method of item 6754, wherein the, composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
6912. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer. 6913. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
6914. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
6915. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
6916. The method of item 6754, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
6917. The method of item 6754 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
6918. The method of item 6754, wherein the device comprises a lubricious coating.
6919. The method of item 6754 wherein the anti-scarring agent is located within pores or holes of the device.
6920. The method of item 6754 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
6921. The method of item 6754, wherein the device comprises a second pharmaceutically active agent.
6922. The method of item 6754 wherein the device comprises an anti-inflammatory agent. 6923. The method of item 6754 wherein the device comprises an agent that inhibits infection.
6924. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
6925. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
6926. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
6927. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
6928. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
6929. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
6930. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
6931. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
6932. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
6933. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
6934. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea. 6935. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
6936. The method of item 6754 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
6937. The method of item 6754, further comprising an antithrombotic agent.
6938. The method of item 6754 wherein the device comprises a visualization agent.
6939. The method of item 6754 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
6940. The method of item 6754 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
6941. The method of item 6754 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
6942. The method of item 6754 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
6943. The method of item 6754 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 6944. The method of item 6754 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
6945. The method of item 6754 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
6946. The method of item 6754 wherein the device comprises an echogenic material.
6947. The method of item 6754 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
6948. The method of item 6754 wherein the device is sterile.
6949. The method of item 6754 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
6950. The method of item 6754 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
6951. The method of item 6754 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
6952. The method of item 6754 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 6953. The method of item 6754 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
6954. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
6955. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
6956. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
6957. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
6958. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
6959. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
6960. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
6961. The method of item 6754 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days. 6962. The method of item 6754 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
6963. The method of item 6754 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
, 6964. The method of item 6754 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
6965. The method of item 6754 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
6966. The method of item 6754 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
6967. The method of item 6754 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6968. The method of item 6754 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6969. The method of item 6754 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6970. The method of item 6754 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6971. The method of item 6754 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 6972. The method of item 6754 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
6973. The method of item 6754 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
6974. The method of item 6754 wherein the combining is performed by spraying the agent or the component onto the sensor.
6975. The method of item 6754 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
6976. The method of item 6754 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
6977. The method of item 6754 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
6978. The method of item 6754 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
6979. The method of item 6754 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
6980. The method of item 6754 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
6981. The method of item 6754 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 6982. The method of item 6754 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
6983. The method of item 6754 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
6984. The method of item 6754 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
6985. The method of item 6754 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
6986. The method of item 6754 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
6987. The method of item 6754 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
6988. The method of item 6754 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
6989. The method of item 6754 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
6990. The method of item 6754 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition. 6991. The method of item 6754 wherein the combining is performed by impregnating the sensor with the agent or the composition.
6992. The method of item 6754 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
6993. The method of item 6754 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
6994. The method of item 6754 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
6995. The method of item 6754 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
6996. The method of item 6754 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
6997. The method of item 6754 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
6998. The method of item 6754 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
6999. The method of item 6754 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor. 7000. The method of item 6754 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
7001. The method of item 6754 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
7002. The method of item 6754 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
7003. The method of item 6754 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
7004. The method of item 6754 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
7005. The method of any one of items 6754-7004 wherein the device monitors blood pressure.
7006. The method of any one of items 6754-7004 wherein the device monitors fluid flow.
7007. The method of any one of items 6754-7004 wherein the device monitors pressure within an aneurysm sac.
7008. The method of any one of items 6754-7004 wherein the device monitors intracranial pressure.
7009. The method of any one of items 6754-7004 wherein the device monitors mechanical pressure associated with a bone fracture. 7010. The method of any one of items 6754-7004 wherein the device monitors barometric pressure.
7011. The method of any one of items 6754-7004 wherein the device monitors eye tremors.
7012. The method of any one of items 6754-7004 wherein the device monitors the depth of a corneal implant.
7013. The method of any one of items 6754-7004 wherein the device monitors intraocular pressure.
7014. The method of any one of items 6754-7004 wherein the device is a passive sensor with an inductor-capacitor circuit.
7015. The method of any one of items 6754-7004 wherein the device is a self-powered strain sensing system.
7016. The method of any one of items 6754-7004 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage.
7017. A method for making a device comprising: combining a cardiac sensor and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
7018. The method of item 7017 wherein the agent is an adensosine A2A receptor antagonist.
7019. The method of item 7017 wherein the agent is an AKT inhibitor.
7020. The method of item 7017 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects
No. 5754 (Merck KgaA). 7021. The method of item 7017 wherein the agent is an alpha 4 integrin antagonist.
7022. The method of item 7017 wherein the agent is an alpha 7 nicotinic receptor agonist.
7023. The method of item 7017 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
7024. The method of item 7017 wherein the agent is an apoptosis antagonist.
7025. The method of item 7017 wherein the agent is an apoptosis activator.
7026. The method of item 7017 wherein the agent is a beta 1 integrin antagonist.
7027. The method of item 7017 wherein the agent is a beta tubulin inhibitor.
7028. The method of item 7017 wherein the agent is a blocker of enzyme production in Hepatitis C.
7029. The method of item 7017 wherein the agent is a Bruton's tyrosine kinase inhibitor.
7030. The method of item 7017 wherein the agent is a calcineurin inhibitor.
7031. The method of item 7017 wherein the agent is a caspase 3 inhibitor.
7032. The method of item 7017 wherein the agent is a CC chemokine receptor antagonist.
7033. The method of item 7017 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 7034. The method of item 7017 wherein the agent is a cathepsin B inhibitor.
7035. The method of item 7017 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), 1NPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7036. The method of item 7017 wherein the agent is a cathepsin L inhibitor.
7037. The method of item 7017 wherein the agent is a CD40 antagonist.
7038. The method of item 7017 wherein the agent is a chemokine receptor agonist.
7039. The method of item 7017 wherein the agent is a chymase inhibitor.
7040. The method of item 7017 wherein the agent is a collagenase antagonist.
7041. The method of item 7017 wherein the agent is a CXCR antagonist.
7042. The method of item 7017 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
7043. The method of item 7017 wherein the agent is a cyclooxygenase 1 inhibitor.
7044. The method of item 7017 wherein the agent is a DHFR inhibitor.
7045. The method of item 7017 wherein the agent is a dual integrin inhibitor.
7046. The method of item 7017 wherein the agent is an elastase inhibitor.
7047. The method of item 7017 wherein the agent is an elongation factor-1 alpha inhibitor.
7048. The method of item 7017 wherein the agent is an endothelial growth factor antagonist.
7049. The method of item 7017 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7050. The method of item 7017 wherein the agent is an endotoxin antagonist. 7051. The method of item 7017 wherein the agent is an epothilone and tubulin binder.
7052. The method of item 7017 wherein the agent is an estrogen receptor antagonist.
7053. The method of item 7017 wherein the agent is an FGF inhibitor.
7054. The method of item 7017 wherein the agent is a famexyl transferase inhibitor.
7055. The method of item 7017 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7056. The method of item 7017 wherein the agent is an FLT-3 kinase inhibitor.
7057. The method of item 7017 wherein the agent is an FGF receptor kinase inhibitor.
7058. The method of item 7017 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201
(Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7059. The method of item 7017 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
7060. The method of item 7017 wherein the agent is a histone deacetylase inhibitor.
7061. The method of item 7017 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
7062. The method of item 7017 wherein the agent is an ICAM inhibitor.
7063. The method of item 7017 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174
(Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
7064. The method of item 7017 wherein the agent is an IL-2 inhibitor.
7065. The method of item 7017 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7066. The method of item 7017 wherein the agent is an IMPDH (inosine monophosphate).
7067. The method of item 7017 wherein the agent is an integrin antagonist.
7068. The method of item 7017 wherein the agent is an interleukin antagonist.
7069. The method of item 7017 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7070. The method of item 7017 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7071. The method of item 7017 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
7072. The method of item 7017 wherein the agent a JAK3 enzyme inhibitor.
7073. The method of item 7017 wherein the agent is a JNK inhibitor.
7074. The method of item 7017 wherein the agent is a kinase inhibitor. 7075. The method of item 7017 wherein the agent is kinesin antagonist.
7076. The method of item 7017 wherein the agent is a kinesin antagonist.
7077. The method of item 7017 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7078. The method of item 7017 wherein the agent is an MAP kinase inhibitor.
7079. The method of item 7017 wherein the agent is a matrix metalloproteinase inhibitor.
7080. The method of item 7017 wherein the agent is an MCP-
CCR2 inhibitor.
7081. The method of item 7017 wherein the agent is an mTOR inhibitor. 7082. The method of item 7017 wherein the agent is an mTOR kinase inhibitor.
7083. The method of item 7017 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
7084. The method of item 7017 wherein the agent is an MIF inhibitor.
7085. The method of item 7017 wherein the agent is an MMP inhibitor.
7086. The method of item 7017 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 7087. The method of item 7017 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
7088. The method of item 7017 wherein the agent is a nitric oxide agonist.
7089. The method of item 7017 wherein the agent is an ornithine decarboxylase inhibitor.
7090. The method of item 7017 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7091. The method of item 7017 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
7092. The method of item 7017 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
7093. The method of item 7017 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glϊmepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7094. The method of item 7017 wherein the agent is a phosphatase inhibitor.
7095. The method of item 7017 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
7096. The method of item 7017 wherein the agent is a PKC inhibitor.
7097. The method of item 7017 wherein the agent is a platelet activating factor antagonist.
7098. The method of item 7017 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
7099. The method of item 7017 wherein the agent is a prolyl hydroxylase inhibitor.
7100. The method of item 7017 wherein the agent is a polymorphonuclear neutrophil inhibitor.
7101. The method of item 7017 wherein the agent is a protein kinase B inhibitor. 7102. The method of item 7017 wherein the agent is a protein kinase C stimulant.
7103. The method of item 7017 wherein the agent is a purine nucleoside analogue.
7104. The method of item 7017 wherein the agent is a purinoreceptor P2X antagonist.
7105. The method of item 7017 wherein the agent is a Raf kinase inhibitor.
7106. The method of item 7017 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7107. The method of item 7017 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7108. The method of item 7017 wherein the agent is an SDF-1 antagonist.
7109. The method of item 7017 wherein the agent is a sheddase inhibitor.
7110. The method of item 7017 wherein the agent is an SRC inhibitor.
7111. The method of item 7017 wherein the agent is a stromelysin inhibitor.
7112. The method of item 7017 wherein the agent is an Syk kinase inhibitor.
7113. The method of item 7017 wherein the agent is a telomerase inhibitor. 7114. The method of item 7017 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
7115. The method of item 7017 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovac.s, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7116. The method of item 7017 wherein the agent is a Toll receptor inhibitor.
7117. The method of item 7017 wherein the agent is a tubulin antagonist.
7118. The method of item 7017 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 7119. The method of item 7017 wherein the agent is a VEGF inhibitor.
7120. The method of item 7017 wherein the agent is a vitamin D receptor agonist.
7121. The method of item 7017 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
7122. The method of item 7017 wherein the agent is AP-23573 (an mTOR inhibitor).
7123. The method of item 7017 wherein the agent is synthadotin (a tubulin antagonist).
7124. The method of item 7017 wherein the agent is S-0885 (a collagenase inhibitor).
7125. The method of item 7017 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
7126. The method of item 7017 wherein the agent is ixabepilone (an epithilone).
7127. The method of item 7017 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
7128. The method of item 7017 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7129. The method of item 7017 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7130. The method of item 7017 wherein the agent is combretastatin (an angiogenesis inhibitor). 7131. The method of item 7017 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7132. The method of item 7017 wherein the agent is SB- 715992 (a kinesin antagonist).
7133. The method of item 7017 wherein the agent is temsirolimus (an mTOR inhibitor).
7134. The method of item 7017 wherein the agent is adalimumab (a TNFα antagonist).
7135. The method of item 7017, wherein the composition comprises a polymer.
7136. The method of item 7017, wherein the composition comprises a polymeric carrier.
7137. The method of item 7017 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
7138. The method of item 7017 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
7139. The method of item 7017 wherein the device has a coating that comprises the anti-scarring agent.
7140. The method of item 7017, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
7141. The method of item 7017, wherein the device has a coating that comprises the agent and directly contacts the sensor.
7142. The method of item 7017, wherein the device has a coating that comprises the agent and indirectly contacts the sensor. 7143. The method of item 7017, wherein the device has a coating that comprises the agent and partially covers the sensor.
7144. The method of item 7017, wherein the device has a coating that comprises the agent and completely covers the sensor.
7145. The method of item 7017, wherein the device has a uniform coating.
7146. The method of item 7017, wherein the device has a nonuniform coating.
7147. The method of item 7017, wherein the device has a discontinuous coating.
7148. The method of item 7017, wherein the device has a patterned coating.
7149. The method of item 7017, wherein the device has a coating with a thickness of 100 μm or less.
7150. The method of item 7017, wherein the device has a coating with a thickness of 10 μm or less.
7151. The method of item 7017, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
7152. The method of item 7017, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
7153. The method of item 7017, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 7154. The method of item 7017, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
7155. The method of item 7017, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
7156. The method of item 7017, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
7157. The method of item 7017, wherein the device has a coating, and wherein the coating further comprises a polymer.
7158. The method of item 7017, wherein the device has a first coating having a first composition and a second coating having a second composition.
7159. The method of item 7017, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7160. The method of item 7017, wherein the composition comprises a polymer.
7161. The method of item 7017, wherein the composition comprises a polymeric carrier.
7162. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer. 7163. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
7164. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
7165. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
7166. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
7167. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
7168. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
7169. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
7170. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
7171. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 7172. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
7173. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
7174. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
7175. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
7176. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
7177. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
7178. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
7179. The method of item 7017, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
7180. The method of item 7017 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer. 7181. The method of item 7017, wherein the device comprises a lubricious coating.
7182. The method of item 7017 wherein the anti-scarring agent is located within pores or holes of the device.
7183. The method of item 7017 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
7184. The method of item 7017, wherein the device comprises a second pharmaceutically active agent.
7185. The method of item 7017 wherein the device comprises an anti-inflammatory agent.
7186. The method of item 7017 wherein the device comprises an agent that inhibits infection.
7187. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
7188. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
7189. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
7190. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
7191. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
7192. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist. 7193. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
7194. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
7195. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
7196. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
7197. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
7198. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
7199. The method of item 7017 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
7200. The method of item 7017, further comprising an antithrombotic agent.
7201. The method of item 7017 wherein the device comprises a visualization agent.
7202. The method of item 7017 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
7203. The method of item 7017 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 7204. The method of item 7017 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
7205. The method of item 7017 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
7206. The method of item 7017 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
7207. The method of item 7017 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
7208. The method of item 7017 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
7209. The method of item 7017 wherein the device comprises an echogenic material.
7210. The method of item 7017 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
7211. The method of item 7017 wherein the device is sterile.
7212. The method of item 7017 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. 7213. The method of item 7017 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
7214. The method of item 7017 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
7215. The method of item 7017 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
7216. The method of item 7017 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
7217. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
7218. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
7219. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
7220. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
7221. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 7222. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
7223. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
7224. The method of item 7017 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
7225. The method of item 7017 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
7226. The method of item 7017 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
7227. The method of item 7017 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
7228. The method of item 7017 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
7229. The method of item 7017 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
7230. The method of item 7017 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7231. The method of item 7017 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 7232. The method of item 7017 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7233. The method of item 7017 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7234. The method of item 7017 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7235. The method of item 7017 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7236. The method of item 7017 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
7237. The method of item 7017 wherein the combining is performed by spraying the agent or the component onto the sensor.
7238. The method of item 7017 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
7239. The method of item 7017 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
7240. The method of item 7017 wherein the combining is performed by covalently attaching the agent or the composition to the sensor. 7241. The method of item 7017 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
7242. The method of item 7017 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
7243. The method of item 7017 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
7244. The method of item 7017 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
7245. The method of item 7017 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
7246. The method of item 7017 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
7247. The method of item 7017 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
7248. The method of item 7017 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
7249. The method of item 7017 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition. 7250. The method of item 7017 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
7251. The method of item 7017 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
7252. The method of item 7017 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
7253. The method of item 7017 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
7254. The method of item 7017 wherein the combining is performed by impregnating the sensor with the agent or the composition.
7255. The method of item 7017 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
7256. The method of item 7017 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
7257. The method of item 7017 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
7258. The method of item 7017 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor. 7259. The method of item 7017 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
7260. The method of item 7017 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
7261. The method of item 7017 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
7262. The method of item 7017 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
7263. The method of item 7017 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
7264. The method of item 7017 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
7265. The method of item 7017 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
7266. The method of item 7017 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
7267. The method of item 7017 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 7268. The method of any one of items 7017-7267 wherein the device monitors cardiac output.
7269. The method of any one of items 7017-7267 wherein the device monitors ejection fraction.
7270. The method of any one of items 7017-7267 wherein the device monitors blood pressure in a heart chamber.
7271. The method of any one of items 7017-7267 wherein the device monitors ventricular wall motions.
7272. The method of any one of items 7017-7267 wherein the device monitors blood flow to a transplanted organ.
7273. The method of any one of items 7017-7267 wherein the device monitors heart rate.
7274. A method for making a device comprising: combining a respiratory sensor and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
7275. The method of item 7274 wherein the agent is an adensosine A2A receptor antagonist.
7276. The method of item 7274 wherein the agent is an AKT inhibitor.
7277. The method of item 7274 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
7278. The method of item 7274 wherein the agent is an alpha 4 integrin antagonist. 7279. The method of item 7274 wherein the agent is an alpha 7 nicotinic receptor agonist.
7280. The method of item 7274 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGlOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
7281. The method of item 7274 wherein the agent is an apoptosis antagonist.
7282. The method of item 7274 wherein the agent is an apoptosis activator.
7283. The method of item 7274 wherein the agent is a beta 1 integrin antagonist.
7284. The method of item 7274 wherein the agent is a beta tubulin inhibitor.
7285. The method of item 7274 wherein the agent is a blocker of enzyme production in Hepatitis C.
7286. The method of item 7274 wherein the agent is a Bruton's tyrosine kinase inhibitor.
7287. The method of item 7274 wherein the agent is a calcineurin inhibitor.
7288. The method of item 7274 wherein the agent is a caspase 3 inhibitor.
7289. The method of item 7274 wherein the agent is a CC chemokine receptor antagonist.
7290. The method of item 7274 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
7291. The method of item 7274 wherein the agent is a cathepsin B inhibitor. 7292. The method of item 7274 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7293. The method of item 7274 wherein the agent is a cathepsin L inhibitor.
7294. The method of item 7274 wherein the agent is a CD40 antagonist.
7295. The method of item 7274 wherein the agent is a chemokine receptor agonist.
7296. The method of item 7274 wherein the agent is a chymase inhibitor.
7297. The method of item 7274 wherein the agent is a collagenase antagonist.
7298. The method of item 7274 wherein the agent is a CXCR antagonist.
7299. The method of item 7274 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a SeπThr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 7300. The method of item 7274 wherein the agent is a cyclooxygenase 1 inhibitor.
7301. The method of item 7274 wherein the agent is a DHFR inhibitor.
7302. The method of item 7274 wherein the agent is a dual integrin inhibitor.
7303. The method of item 7274 wherein the agent is an elastase inhibitor.
7304. The method of item 7274 wherein the agent is an elongation factor- 1 alpha inhibitor.
7305. The method of item 7274 wherein the agent is an endothelial growth factor antagonist.
7306. The method of item 7274 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7307. The method of item 7274 wherein the agent is an endotoxin antagonist.
7308. The method of item 7274 wherein the agent is an epothilone and tubulin binder. 7309. The method of item 7274 wherein the agent is an estrogen receptor antagonist.
7310. The method of item 7274 wherein the agent is an FGF inhibitor.
7311. The method of item 7274 wherein the agent is a famexyl transferase inhibitor.
7312. The method of item 7274 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme),
Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7313. The method of item 7274 wherein the agent is an FLT-3 kinase inhibitor.
7314. The method of item 7274 wherein the agent is an FGF receptor kinase inhibitor.
7315. The method of item 7274 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7316. The method of item 7274 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
7317. The method of item 7274 wherein the agent is a histone deacetylase inhibitor.
7318. The method of item 7274 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
7319. The method of item 7274 wherein the agent is an ICAM inhibitor.
7320. The method of item 7274 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
7321. The method of item 7274 wherein the agent is an IL-2 inhibitor.
7322. The method of item 7274 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (lnflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7323. The method of item 7274 wherein the agent is an IMPDH (inosine monophosphate).
7324. The method of item 7274 wherein the agent is an integrin antagonist.
7325. The method of item 7274 wherein the agent is an interleukin antagonist.
7326. The method of item 7274 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7327. The method of item 7274 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7328. The method of item 7274 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
7329. The method of item 7274 wherein the agent a JAK3 enzyme inhibitor.
7330. The method of item 7274 wherein the agent is a JNK inhibitor.
7331. The method of item 7274 wherein the agent is a kinase inhibitor. 7332. The method of item 7274 wherein the agent is kinesin antagonist.
7333. The method of item 7274 wherein the agent is a kinesin antagonist.
7334. The method of item 7274 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7335. The method of item 7274 wherein the agent is an MAP kinase inhibitor.
7336. The method of item 7274 wherein the agent is a matrix metalloproteinase inhibitor.
7337. The method of item 7274 wherein the agent is an MCP-
CCR2 inhibitor.
7338. The method of item 7274 wherein the agent is an mTOR inhibitor. 7339. The method of item 7274 wherein the agent is an mTOR kinase inhibitor.
7340. The method of item 7274 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
7341. The method of item 7274 wherein the agent is an MIF inhibitor.
7342. The method of item 7274 wherein the agent is an MMP inhibitor.
7343. The method of item 7274 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 7344. The method of item 7274 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
7345. The method of item 7274 wherein the agent is a nitric oxide agonist.
7346. The method of item 7274 wherein the agent is an ornithine decarboxylase inhibitor.
7347. The method of item 7274 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7348. The method of item 7274 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
7349. The method of item 7274 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering'AG), and an analogue or derivative thereof.
7350. The method of item 7274 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKϋne), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7351. The method of item 7274 wherein the agent is a phosphatase inhibitor.
7352. The method of item 7274 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
7353. The method of item 7274 wherein the agent is a PKC inhibitor.
7354. The method of item 7274 wherein the agent is a platelet activating factor antagonist.
7355. The method of item 7274 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
7356. The method of item 7274 wherein the agent is a prolyl hydroxylase inhibitor.
7357. The method of item 7274 wherein the agent is a polymorphonuclear neutrophil inhibitor.
7358. The method of item 7274 wherein the agent is a protein kinase B inhibitor. 7359. The method of item 7274 wherein the agent is a protein kinase C stimulant.
7360. The method of item 7274 wherein the agent is a purine nucleoside analogue.
7361. The method of item 7274 wherein the agent is a purinoreceptor P2X antagonist.
7362. The method of item 7274 wherein the agent is a Raf kinase inhibitor.
7363. The method of item 7274 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7364. The method of item 7274 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7365. The method of item 7274 wherein the agent is an SDF-1 antagonist.
7366. The method of item 7274 wherein the agent is a sheddase inhibitor.
7367. The method of item 7274 wherein the agent is an SRC inhibitor.
7368. The method of item 7274 wherein the agent is a stromelysin inhibitor.
7369. The method of item 7274 wherein the agent is an Syk kinase inhibitor.
7370. The method of item 7274 wherein the agent is a telomerase inhibitor. 7371. The method of item 7274 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
7372. The method of item 7274 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7373. The method of item 7274 wherein the agent is a Toll receptor inhibitor.
7374. The method of item 7274 wherein the agent is a tubulin antagonist.
7375. The method of item 7274 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4)- (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 7376. The method of item 7274 wherein the agent is a VEGF inhibitor.
7377. The method of item 7274 wherein the agent is a vitamin D receptor agonist.
7378. The method of item 7274 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
7379. The method of item 7274 wherein the agent is AP-23573 (an mTOR inhibitor).
7380. The method of item 7274 wherein the agent is synthadotin (a tubulin antagonist).
7381. The method of item 7274 wherein the agent is S-0885 (a collagenase inhibitor).
7382. The method of item 7274 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
7383. The method of item 7274 wherein the agent is ixabepilone (an epithilone).
7384. The method of item 7274 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
7385. The method of item 7274 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7386. The method of item 7274 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7387. The method of item 7274 wherein the agent is combretastatin (an angiogenesis inhibitor). 7388. The method of item 7274 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7389. The method of item 7274 wherein the agent is SB- 715992 (a kinesin antagonist).
7390. The method of item 7274 wherein the agent is temsirolimus (an mTOR inhibitor).
7391. The method of item 7274 wherein the agent is adalimumab (a TNFα antagonist).
7392. The method of item 7274, wherein the composition comprises a polymer.
7393. The method of item 7274, wherein the composition comprises a polymeric, carrier.
7394. The method of item 7274 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
7395. The method of item 7274 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
7396. The method of item 7274 wherein the device has a coating that comprises the anti-scarring agent.
7397. The method of item 7274, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
7398. The method of item 7274, wherein the device has a coating that comprises the agent and directly contacts the sensor.
7399. The method of item 7274, wherein the device has a coating that comprises the agent and indirectly contacts the sensor. 7400. The method of item 7274, wherein the device has a coating that comprises the agent and partially covers the sensor.
7401. The method of item 7274, wherein the device has a coating that comprises the agent and completely covers the sensor.
7402. The method of item 7274, wherein the device has a uniform coating.
7403. The method of item 7274, wherein the device has a nonuniform coating.
7404. The method of item 7274, wherein the device has a discontinuous coating.
7405. The method of item 7274, wherein the device has a patterned coating.
7406. The method of item 7274, wherein the device has a coating with a thickness of 100 μm or less.
7407. The method of item 7274, wherein the device has a coating with a thickness of 10 μm or less.
7408. The method of item 7274, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
7409. The method of item 7274, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
7410. The method of item 7274, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 7411. The method of item 7274, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
7412. The method of item 7274, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
7413. The method of item 7274, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
7414. The method of item 7274, wherein the device has a coating, and wherein the coating further comprises a polymer.
7415. The method of item 7274, wherein the device has a first coating having a first composition and a second coating having a second composition.
7416. The method of item 7274, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7417. The method of item 7274, wherein the composition comprises a polymer.
7418. The method of item 7274, wherein the composition comprises a polymeric carrier.
7419. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer. 7420. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
7421. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
7422. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
7423. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
7424. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
7425. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
7426. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
7427. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
7428. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 7429. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
7430. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
7431. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
7432. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
7433. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
7434. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
7435. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
7436. The method of item 7274, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
7437. The method of item 7274 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer. 7438. The method of item 7274, wherein the device comprises a lubricious coating.
7439. The method of item 7274 wherein the anti-scarring agent is located within pores or holes of the device.
7440. The method of item 7274 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
7441. The method of item 7274, wherein the device comprises a second pharmaceutically active agent.
7442. The method of item 7274 wherein the device comprises an anti-inflammatory agent.
7443. The method of item 7274 wherein the device comprises an agent that inhibits infection.
7444. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
7445. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
7446. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
7447. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
7448. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
7449. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist. 7450. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
7451. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
7452. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
7453. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
7454. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
7455. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
7456. The method of item 7274 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
7457. The method of item 7274, further comprising an antithrombotic agent.
7458. The method of item 7274 wherein the device comprises a visualization agent.
7459. The method of item 7274 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
7460. The method of item 7274 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 7461. The method of item 7274 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
7462. The method of item 7274 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
7463. The method of item 7274 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
7464. The method of item 7274 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
7465. The method of item 7274 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
7466. The method of item 7274 wherein the device comprises an echogenic material.
7467. The method of item 7274 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
7468. The method of item 7274 wherein the device is sterile.
7469. The method of item 7274 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. 7470. The method of item 7274 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
7471. The method of item 7274 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
7472. The method of item 7274 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
7473. The method of item 7274 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
7474. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
7475. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
7476. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
7477. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
7478. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 7479. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
7480. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
7481. The method of item 7274 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
7482. The method of item 7274 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
7483. The method of item 7274 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
7484. The method of item 7274 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
7485. The method of item 7274 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
7486. The method of item 7274 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
7487. The method of item 7274 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7488. The method of item 7274 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 7489. The method of item 7274 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7490. The method of item 7274 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7491. The method of item 7274 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7492. The method of item 7274 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7493. The method of item 7274 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
7494. The method of item 7274 wherein the combining is performed by spraying the agent or the component onto the sensor.
7495. The method of item 7274 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
7496. The method of item 7274 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
7497. The method of item 7274 wherein the combining is performed by covalently attaching the agent or the composition to the sensor. 7498. The method of item 7274 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
7499. The method of item 7274 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
7500. The method of item 7274 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
7501. The method of item 7274 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
7502. The method of item 7274 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
7503. The method of item 7274 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
7504. The method of item 7274 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
7505. The method of item 7274 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
7506. The method of item 7274 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition. 7507. The method of item 7274 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
7508. The method of item 7274 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
7509. The method of item 7274 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
7510. The method of item 7274 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
7511. The method of item 7274 wherein the combining is performed by impregnating the sensor with the agent or the composition.
7512. The method of item 7274 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
7513. The method of item 7274 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
7514. The method of item 7274 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
7515. The method of item 7274 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor. 7516. The method of item 7274 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
7517. The method of item 7274 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
7518. The method of item 7274 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
7519. The method of item 7274 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
7520. The method of item 7274 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
7521. The method of item 7274 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
7522. The method of item 7274 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
7523. The method of item 7274 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
7524. The method of item 7274 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 7525. The method of any one of items 7274-7524 wherein the device monitors pulmonary functions.
7526. A method for making a device comprising: combining an auditory sensor and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
7527. The method of item 7526 wherein the agent is an adensosine A2A receptor antagonist.
7528. The method of item 7526 wherein the agent is an AKT inhibitor.
7529. The method of item 7526 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
7530. The method of item 7526 wherein the agent is an alpha 4 integrin antagonist.
7531. The method of item 7526 wherein the agent is an alpha 7 nicotinic receptor agonist.
7532. The method of item 7526 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG)1 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
7533. The method of item 7526 wherein the agent is an apoptosis antagonist.
7534. The method of item 7526 wherein the agent is an apoptosis activator.
7535. The method of item 7526 wherein the agent is a beta 1 integrin antagonist.
7536. The method of item 7526 wherein the agent is a beta tubulin inhibitor. 7537. The method of item 7526 wherein the agent is a blocker of enzyme production in Hepatitis C.
7538. The method of item 7526 wherein the agent is a Bruton's tyrosine kinase inhibitor.
7539. The method of item 7526 wherein the agent is a calcineurin inhibitor.
7540. The method of item 7526 wherein the agent is a caspase 3 inhibitor.
7541. The method of item 7526 wherein the agent is a CC chemokine receptor antagonist.
7542. The method of item 7526 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
7543. The method of item 7526 wherein the agent is a cathepsin B inhibitor.
7544. The method of item 7526 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7545. The method of item 7526 wherein the agent is a cathepsin L inhibitor.
7546. The method of item 7526 wherein the agent is a CD40 antagonist.
7547. The method of item 7526 wherein the agent is a chemokine receptor agonist. 7548. The method of item 7526 wherein the agent is a chymase inhibitor.
7549. The method of item 7526 wherein the agent is a collagenase antagonist.
7550. The method of item 7526 wherein the agent is a CXCR antagonist.
7551. The method of item 7526 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
7552. The method of item 7526 wherein the agent is a cyclooxygenase 1 inhibitor.
7553. The method of item 7526 wherein the agent is a DHFR inhibitor.
7554. The method of item 7526 wherein the agent is a dual integrin inhibitor.
7555. The method of item 7526 wherein the agent is an elastase inhibitor.
7556. The method of item 7526 wherein the agent is an elongation factor-1 alpha inhibitor. 7557. The method of item 7526 wherein the agent is an endothelial growth factor antagonist.
7558. The method of item 7526 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7559. The method of item 7526 wherein the agent is an endotoxin antagonist.
7560. The method of item 7526 wherein the agent is an epothilone and tubulin binder.
7561. The method of item 7526 wherein the agent is an estrogen receptor antagonist.
7562. The method of item 7526 wherein the agent is an FGF inhibitor.
7563. The method of item 7526 wherein the agent is a famexyl transferase inhibitor.
7564. The method of item 7526 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7565. The method of item 7526 wherein the agent is an FLT-3 kinase inhibitor.
7566. The method of item 7526 wherein the agent is an FGF receptor kinase inhibitor.
7567. The method of item 7526 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7568. The method of item 7526 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
7569. The method of item 7526 wherein the agent is a histone deacetylase inhibitor.
7570. The method of item 7526 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 7571. The method of item 7526 wherein the agent is an ICAM inhibitor.
7572. The method of item 7526 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
7573. The method of item 7526 wherein the agent is an IL-2 inhibitor.
7574. The method of item 7526 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7575. The method of item 7526 wherein the agent is an IMPDH (inosine monophosphate). 7576. The method of item 7526 wherein the agent is an integrin antagonist.
7577. The method of item 7526 wherein the agent is an interleukin antagonist.
7578. The method of item 7526 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7579. The method of item 7526 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7580. The method of item 7526 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
7581. The method of item 7526 wherein the agent a JAK3 enzyme inhibitor.
7582. The method of item 7526 wherein the agent is a JNK inhibitor.
7583. The method of item 7526 wherein the agent is a kinase inhibitor.
7584. The method of item 7526 wherein the agent is kinesin antagonist.
7585. The method of item 7526 wherein the agent is a kinesin antagonist.
7586. The method of item 7526 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7587. The method of item 7526 wherein the agent is an MAP kinase inhibitor.
7588. The method of item 7526 wherein the agent is a matrix metalloproteinase inhibitor.
7589. The method of item 7526 wherein the agent is an MCP-
CCR2 inhibitor.
7590. The method of item 7526 wherein the agent is an mTOR inhibitor.
7591. The method of item 7526 wherein the agent is an mTOR kinase inhibitor.
7592. The method of item 7526 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
7593. The method of item 7526 wherein the agent is an MIF inhibitor.
7594. The method of item 7526 wherein the agent is an MMP inhibitor.
7595. The method of item 7526 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
7596. The method of item 7526 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 7597. The method of item 7526 wherein the agent is a nitric oxide agonist.
7598. The method of item 7526 wherein the agent is an ornithine decarboxylase inhibitor.
7599. The method of item 7526 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7600. The method of item 7526 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
7601. The method of item 7526 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
7602. The method of item 7526 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7603. The method of item 7526 wherein the agent is a phosphatase inhibitor.
7604. The method of item 7526 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
7605. The method of item 7526 wherein the agent is a PKC inhibitor.
7606. The method of item 7526 wherein the agent is a platelet activating factor antagonist.
7607. The method of item 7526 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
7608. The method of item 7526 wherein the agent is a prolyl hydroxylase inhibitor.
7609. The method of item 7526 wherein the agent is a polymorphonuclear neutrophil inhibitor.
7610. The method of item 7526 wherein the agent is a protein kinase B inhibitor.
7611. The method of item 7526 wherein the agent is a protein kinase C stimulant.
7612. The method of item 7526 wherein the agent is a purine nucleoside analogue.
7613. The method of item 7526 wherein the agent is a purinoreceptor P2X antagonist.
7614. The method of item 7526 wherein the agent is a Raf kinase inhibitor. 7615. The method of item 7526 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7616. The method of item 7526 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7617. The method of item 7526 wherein the agent is an SDF-1 antagonist.
7618. The method of item 7526 wherein the agent is a sheddase inhibitor.
7619. The method of item 7526 wherein the agent is an SRC inhibitor.
7620. The method of item 7526 wherein the agent is a stromelysin inhibitor.
7621. The method of item 7526 wherein the agent is an Syk kinase inhibitor.
7622. The method of item 7526 wherein the agent is a telomerase inhibitor.
7623. The method of item 7526 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
7624. The method of item 7526 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7625. The method of item 7526 wherein the agent is a Toll receptor inhibitor.
7626. The method of item 7526 wherein the agent is a tubulin antagonist.
7627. The method of item 7526 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
7628. The method of item 7526 wherein the agent is a VEGF inhibitor.
7629. The method of item 7526 wherein the agent is a vitamin D receptor agonist.
7630. The method of item 7526 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
7631. The method of item 7526 wherein the agent is AP-23573 (an mTOR inhibitor). 7632. The method of item 7526 wherein the agent is synthadotin (a tubulin antagonist).
7633. The method of item 7526 wherein the agent is S-0885 (a collagenase inhibitor).
7634. The method of item 7526 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
7635. The method of item 7526 wherein the agent is ixabepilone (an epithilone).
7636. The method of item 7526 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
7637. The method of item 7526 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7638. The method of item 7526 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7639. The method of item 7526 wherein the agent is combretastatin (an angiogenesis inhibitor).
7640. The method of item 7526 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7641. The method of item 7526 wherein the agent is SB- 715992 (a kinesin antagonist).
7642. The method of item 7526 wherein the agent is temsirolimus (an mTOR inhibitor).
7643. The method of item 7526 wherein the agent is adalimumab (a TNFα antagonist). 7644. The method of item 7526, wherein the composition comprises a polymer.
7645. The method of item 7526, wherein the composition comprises a polymeric carrier.
7646. The method of item 7526 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
7647. The method of item 7526 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
7648. The method of item 7526 wherein the device has a coating that comprises the anti-scarring agent.
7649. The method of item 7526, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
7650. The method of item 7526, wherein the device has a coating that comprises the agent and directly contacts the sensor.
7651. The method of item 7526, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
7652. The method of item 7526, wherein the device has a coating that comprises the agent and partially covers the sensor.
7653. The method of item 7526, wherein the device has a coating that comprises the agent and completely covers the sensor.
7654. The method of item 7526, wherein the device has a uniform coating.
7655. The method of item 7526, wherein the device has a non- uniform coating. 7656. The method of item 7526, wherein the device has a discontinuous coating.
7657. The method of item 7526, wherein the device has a patterned coating.
7658. The method of item 7526, wherein the device has a coating with a thickness of 100 μm or less.
7659. The method of item 7526, wherein the device has a coating with a thickness of 10 μm or less.
7660. The method of item 7526, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
7661. The method of item 7526, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
7662. The method of item 7526, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
7663. The method of item 7526, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
7664. The method of item 7526, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
7665. The method of item 7526, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 7666. The method of item 7526, wherein the device has a coating, and wherein the coating further comprises a polymer.
7667. The method of item 7526, wherein the device has a first coating having a first composition and a second coating having a second composition.
7668. The method of item 7526, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7669. The method of item 7526, wherein the composition comprises a polymer.
7670. The method of item 7526, wherein the composition comprises a polymeric carrier.
7671. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
7672. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
7673. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
7674. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 7675. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
7676. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
7677. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
7678. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
7679. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
7680. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
7681. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
7682. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
7683. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 7684. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
7685. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
7686. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
7687. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
7688. The method of item 7526, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
7689. The method of item 7526 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
7690. The method of item 7526, wherein the device comprises a lubricious coating.
7691. The method of item 7526 wherein the anti-scarring agent is located within pores or holes of the device.
7692. The method of item 7526 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
7693. The method of item 7526, wherein the device comprises a second pharmaceutically active agent. 7694. The method of item 7526 wherein the device comprises an anti-inflammatory agent.
7695. The method of item 7526 wherein the device comprises an agent that inhibits infection.
7696. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
7697. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
7698. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
7699. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
7700. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
7701. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
7702. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
7703. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
7704. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
7705. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 7706. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
7707. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
7708. The method of item 7526 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
7709. The method of item 7526, further comprising an antithrombotic agent.
7710. The method of item 7526 wherein the device comprises a visualization agent
7711. The method of item 7526 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
7712. The method of item 7526 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
7713. The method of item 7526 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
7714. The method of item 7526 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 7715. The method of item 7526 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
7716. The method of item 7526 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
7717. The method of item 7526 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
7718. The method of item 7526 wherein the device comprises an echogenic material.
7719. The method of item 7526 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
7720. The method of item 7526 wherein the device is sterile.
7721. The method of item 7526 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
7722. The method of item 7526 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
7723. The method of item 7526 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 7724. The method of item 7526 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
7725. The method of item 7526 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
7726. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
7727. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
7728. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
7729. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
7730. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
7731. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
7732. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 7733. The method of item 7526 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
7734. The method of item 7526 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
7735. The method of item 7526 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
7736. The method of item 7526 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
7737. The method of item 7526 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
7738. The method of item 7526 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
7739. The method of item 7526 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7740. The method of item 7526 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7741. The method of item 7526 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7742. The method of item 7526 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 7743. The method of item 7526 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7744. The method of item 7526 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7745. The method of item 7526 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
7746. The method of item 7526 wherein the combining is performed by spraying the agent or the component onto the sensor.
7747. The method of item 7526 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
7748. The method of item 7526 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
7749. The method of item 7526 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
7750. The method of item 7526 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
7751. The method of item 7526 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
7752. The method of item 7526 wherein the combining is performed by coating the sensor with a substance that absorbs the agent. 7753. The method of item 7526 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
7754. The method of item 7526 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
7755. The method of item 7526 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
7756. The method of item 7526 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
7757. The method of item 7526 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
7758. The method of item 7526 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
7759. The method of item 7526 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
7760. The method of item 7526 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
7761. The method of item 7526 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition. 7762. The method of item 7526 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
7763. The method of item 7526 wherein the combining is performed by impregnating the sensor with the agent or the composition.
7764. The method of item 7526 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
7765. The method of item 7526 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
7766. The method of item 7526 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
7767. The method of item 7526 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
7768. The method of item 7526 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
7769. The method of item 7526 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
7770. The method of item 7526 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 7771. The method of item 7526 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
7772. The method of item 7526 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
7773. The method of item 7526 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
7774. The method of item 7526 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
7775. The method of item 7526 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
7776. The method of item 7526 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
7777. The method of any one of items 7526-7776 wherein the device is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
7778. The method of any one of items 7526-7776 wherein the device generates an electrical audio signal.
7779. The method of any one of items 7526-7776 wherein the device is a capacitive sensor that is coupled to a vibrating auditory element. 7780. The method of any one of items 7526-7776 wherein the device is an electromagnetic sensor.
7781. A method for making a device comprising: combining an electrolyte or metabolite sensor and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
7782. The method of item 7781 wherein the agent is an adensosine A2A receptor antagonist.
7783. The method of item 7781 wherein the agent is an AKT inhibitor.
7784. The method of item 7781 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
7785. The method of item 7781 wherein the agent is an alpha 4 integrin antagonist.
7786. The method of item 7781 wherein the agent is an alpha 7 nicotinic receptor agonist.
7787. The method of item 7781 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
7788. The method of item 7781 wherein the agent is an apoptosis antagonist.
7789. The method of item 7781 wherein the agent is an apoptosis activator.
7790. The method of item 7781 wherein the agent is a beta 1 integrin antagonist.
7791. The method of item 7781 wherein the agent is a beta tubulin inhibitor. 7792. The method of item 7781 wherein the agent is a blocker of enzyme production in Hepatitis C.
7793. The method of item 7781 wherein the agent is a Bruton's tyrosine kinase inhibitor.
7794. The method of item 7781 wherein the agent is a calcineurin inhibitor.
7795. The method of item 7781 wherein the agent is a caspase 3 inhibitor.
7796. The method of item 7781 wherein the agent is a CC chemokine receptor antagonist.
7797. The method of item 7781 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
7798. The method of item 7781 wherein the agent is a cathepsin B inhibitor.
7799. The method of item 7781 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7800. The method of item 7781 wherein the agent is a cathepsin L inhibitor.
7801. The method of item 7781 wherein the agent is a CD40 antagonist.
7802. The method of item 7781 wherein the agent is a chemokine receptor agonist. 7803. The method of item 7781 wherein the agent is a chymase inhibitor.
7804. The method of item 7781 wherein the agent is a collagenase antagonist.
7805. The method of item 7781 wherein the agent is a CXCR antagonist.
7806. The method of item 7781 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
7807. The method of item 7781 wherein the agent is a cyclooxygenase 1 inhibitor.
7808. The method of item 7781 wherein the agent is a DHFR inhibitor.
7809. The method of item 7781 wherein the agent is a dual integrin inhibitor.
7810. The method of item 7781 wherein the agent is an elastase inhibitor.
7811. The method of item 7781 wherein the agent is an elongation factor-1 alpha inhibitor. 7812. The method of item 7781 wherein the agent is an endothelial growth factor antagonist.
7813. The method of item 7781 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7814. The method of item 7781 wherein the agent is an endotoxin antagonist.
7815. The method of item 7781 wherein the agent is an epothilone and tubulin binder.
7816. The method of item 7781 wherein the agent is an estrogen receptor antagonist.
7817. The method of item 7781 wherein the agent is an FGF inhibitor.
7818. The method of item 7781 wherein the agent is a farnexyl transferase inhibitor.
7819. The method of item 7781 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7820. The method of item 7781 wherein the agent is an FLT-3 kinase inhibitor.
7821. The method of item 7781 wherein the agent is an FGF receptor kinase inhibitor.
7822. The method of item 7781 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7823. The method of item 7781 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, T,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
7824. The method of item 7781 wherein the agent is a histone deacetylase inhibitor.
7825. The method of item 7781 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 7826. The method of item 7781 wherein the agent is an ICAM inhibitor.
7827. The method of item 7781 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
7828. The method of item 7781 wherein the agent is an IL-2 inhibitor.
7829. The method of item 7781 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7830. The method of item 7781 wherein the agent is an IMPDH (inosine monophosphate). 7831. The method of item 7781 wherein the agent is an integrin antagonist.
7832. The method of item 7781 wherein the agent is an interleukin antagonist.
7833. The method of item 7781 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7834. The method of item 7781 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7835. The method of item 7781 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
7836. The method of item 7781 wherein the agent a JAK3 enzyme inhibitor.
7837. The method of item 7781 wherein the agent is a JNK inhibitor.
7838. The method of item 7781 wherein the agent is a kinase inhibitor.
7839. The method of item 7781 wherein the agent is kinesin antagonist.
7840. The method of item 7781 wherein the agent is a kinesin antagonist.
7841. The method of item 7781 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7842. The method of item 7781 wherein the agent is an MAP kinase inhibitor.
7843. The method of item 7781 wherein the agent is a matrix metalloproteinase inhibitor.
7844. The method of item 7781 wherein the agent is an MCP-
CCR2 inhibitor.
7845. The method of item 7781 wherein the agent is an mTOR inhibitor.
7846. The method of item 7781 wherein the agent is an mTOR kinase inhibitor.
7847. The method of item 7781 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
7848. The method of item 7781 wherein the agent is an MIF inhibitor.
7849. The method of item 7781 wherein the agent is an MMP inhibitor.
7850. The method of item 7781 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
7851. The method of item 7781 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 7852. The method of item 7781 wherein the agent is a nitric oxide agonist.
7853. The method of item 7781 wherein the agent is an ornithine decarboxylase inhibitor.
7854. The method of item 7781 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7855. The method of item 7781 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
7856. The method of item 7781 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
7857. The method of item 7781 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7858. The method of item 7781 wherein the agent is a phosphatase inhibitor.
7859. The method of item 7781 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
7860. The method of item 7781 wherein the agent is a PKC inhibitor.
7861. The method of item 7781 wherein the agent is a platelet activating factor antagonist.
7862. The method of item 7781 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
7863. The method of item 7781 wherein the agent is a prolyl hydroxylase inhibitor.
7864. The method of item 7781 wherein the agent is a polymorphonuclear neutrophil inhibitor.
7865. The method of item 7781 wherein the agent is a protein kinase B inhibitor.
7866. The method of item 7781 wherein the agent is a protein kinase C stimulant.
7867. The method of item 7781 wherein the agent is a purine nucleoside analogue.
7868. The method of item 7781 wherein the agent is a purinoreceptor P2X antagonist.
7869. The method of item 7781 wherein the agent is a Raf kinase inhibitor. 7870. The method of item 7781 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7871. The method of item 7781 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7872. The method of item 7781 wherein the agent is an SDF-1 antagonist.
7873. The method of item 7781 wherein the agent is a sheddase inhibitor.
7874. The method of item 7781 wherein the agent is an SRC inhibitor.
7875. The method of item 7781 wherein the agent is a stromelysin inhibitor.
7876. The method of item 7781 wherein the agent is an Syk kinase inhibitor.
7877. The method of item 7781 wherein the agent is a telomerase inhibitor.
7878. The method of item 7781 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
7879. The method of item 7781 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS' Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7880. The method of item 7781 wherein the agent is a Toll receptor inhibitor.
7881. The method of item 7781 wherein the agent is a tubulin antagonist.
7882. The method of item 7781 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
7883. The method of item 7781 wherein the agent is a VEGF inhibitor.
7884. The method of item 7781 wherein the agent is a vitamin D receptor agonist.
7885. The method of item 7781 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
7886. The method of item 7781 wherein the agent is AP-23573 (an mTOR inhibitor). 7887. The method of item 7781 wherein the agent is synthadotin (a tubulin antagonist).
7888. The method of item 7781 wherein the agent is S-0885 (a collagenase inhibitor).
7889. The method of item 7781 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
7890. The method of item 7781 wherein the agent is ixabepilone (an epithilone).
7891. The method of item 7781 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
7892. The method of item 7781 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7893. The method of item 7781 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7894. The method of item 7781 wherein the agent is combretastatin (an angiogenesis inhibitor).
7895. The method of item 7781 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7896. The method of item 7781 wherein the agent is SB- 715992 (a kinesin antagonist).
7897. The method of item 7781 wherein the agent is temsirolimus (an mTOR inhibitor).
7898. The method of item 7781 wherein the agent is adalimumab (a TNFα antagonist). 7899. The method of item 7781 , wherein the composition comprises a polymer.
7900. The method of item 7781 , wherein the composition comprises a polymeric carrier.
7901. The method of item 7781 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
7902. The method of item 7781 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
7903. The method of item 7781 wherein the device has a coating that comprises the anti-scarring agent. t
7904. The method of item 7781 , wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
7905. The method of item 7781 , wherein the device has a coating that comprises the agent and directly contacts the sensor.
7906. The method of item 7781 , wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
7907. The method of item 7781 , wherein the device has a coating that comprises the agent and partially covers the sensor.
7908. The method of item 7781 , wherein the device has a coating that comprises the agent and completely covers the sensor.
7909. The method of item 7781 , wherein the device has a uniform coating.
7910. The method of item 7781 , wherein the device has a non- uniform coating. 7911. The method of item 7781 , wherein the device has a discontinuous coating.
7912. The method of item 7781 , wherein the device has a patterned coating.
7913. The method of item 7781 , wherein the device has a coating with a thickness of 100 μm or less.
7914. The method of item 7781 , wherein the device has a coating with a thickness of 10 μm or less.
7915. The method of item 7781 , wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
7916. The method of item 7781 , wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
7917. The method of item 7781 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
7918. The method of item 7781 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
7919. The method of item 7781 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
7920. The method of item 7781 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 7921. The method of item 7781 , wherein the device has a coating, and wherein the coating further comprises a polymer.
7922. The method of item 7781 , wherein the device has a first coating having a first composition and a second coating having a second composition.
7923. The method of item 7781 , wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7924. The method of item 7781 , wherein the composition comprises a polymer.
7925. The method of item 7781 , wherein the composition comprises a polymeric carrier.
7926. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
7927. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
7928. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
7929. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 7930. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
7931. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
7932. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
7933. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
7934. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
7935. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
7936. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
7937. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
7938. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 7939. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
7940. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
7941. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
7942. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
7943. The method of item 7781 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
7944. The method of item 7781 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
7945. The method of item 7781 , wherein the device comprises a lubricious coating.
7946. The method of item 7781 wherein the anti-scarring agent is located within pores or holes of the device.
7947. The method of item 7781 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
7948. The method of item 7781 , wherein the device comprises a second pharmaceutically active agent. 7949. The method of item 7781 wherein the device comprises an anti-inflammatory agent.
7950. The method of item 7781 wherein the device comprises an agent that inhibits infection.
7951. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
7952. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
7953. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
7954. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
7955. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
7956. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
7957. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
7958. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
7959. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
7960. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 7961. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
7962. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
7963. The method of item 7781 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
7964. The method of item 7781 , further comprising an antithrombotic agent.
7965. The method of item 7781 wherein the device comprises a visualization agent.
7966. The method of item 7781 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
7967. The method of item 7781 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
7968. The method of item 7781 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
7969. The method of item 7781 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 7970. The method of item 7781 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
7971. The method of item 7781 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
7972. The method of item 7781 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
7973. The method of item 7781 wherein the device comprises an echogenic material.
7974. The method of item 7781 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
7975. The method of item 7781 wherein the device is sterile.
7976. The method of item 7781 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
7977. The method of item 7781 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
7978. The method of item 7781 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 7979. The method of item 7781 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
7980. The method of item 7781 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
7981. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
7982. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
7983. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
7984. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
7985. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
7986. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
7987. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 7988. The method of item 7781 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
7989. The method of item 7781 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
7990. The method of item 7781 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
7991. The method of item 7781 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
7992. The method of item 7781 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
7993. The method of item 7781 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
7994. The method of item 7781 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7995. The method of item 7781 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7996. The method of item 7781 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7997. The method of item 7781 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 7998. The method of item 7781 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
7999. The method of item 7781 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8000. The method of item 7781 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
8001. The method of item 7781 wherein the combining is performed by spraying the agent or the component onto the sensor.
8002. The method of item 7781 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
8003. The method of item 7781 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
8004. The method of item 7781 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
8005. The method of item 7781 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
8006. The method of item 7781 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
8007. The method of item 7781 wherein the combining is performed by coating the sensor with a substance that absorbs the agent. 8008. The method of item 7781 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
8009. The method of item 7781 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
8010. The method of item 7781 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
8011. The method of item 7781 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
8012. The method of item 7781 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
8013. The method of item 7781 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
8014. The method of item 7781 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
8015. The method of item 7781 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
8016. The method of item 7781 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition. 8017. The method of item 7781 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
8018. The method of item 7781 wherein the combining is performed by impregnating the sensor with the agent or the composition.
8019. The method of item 7781 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
8020. The method of item 7781 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
8021. The method of item 7781 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8022. The method of item 7781 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8023. The method of item 7781 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
8024. The method of item 7781 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8025. The method of item 7781 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 8026. The method of item 7781 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
8027. The method of item 7781 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8028. The method of item 7781 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8029. The method of item 7781 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
8030. The method of item 7781 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8031. The method of item 7781 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
8032. The method of any one of items 7781-8031 wherein the device emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
8033. The method of any one of items 7781-8031 wherein the device is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader. 8034. The method of any one of items 7781-8031 wherein the device is a monolithic bioelectronic device for detecting at least one analyte within the host.
8035. A method for making a device comprising: combining a pump and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
8036. The method of item 8035 wherein the agent is an adensosine A2A receptor antagonist.
8037. The method of item 8035 wherein the agent is an AKT inhibitor.
8038. The method of item 8035 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8039. The method of item 8035 wherein the agent is an alpha 4 integrin antagonist.
8040. The method of item 8035 wherein the agent is an alpha 7 nicotinic receptor agonist.
8041. The method of item 8035 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8042. The method of item 8035 wherein the agent is an apoptosis antagonist.
8043. The method of item 8035 wherein the agent is an apoptosis activator.
8044. The method of item 8035 wherein the agent is a beta 1 integrin antagonist.
8045. The method of item 8035 wherein the agent is a beta tubulin inhibitor. 8046. The method of item 8035 wherein the agent is a blocker of enzyme production in Hepatitis C.
8047. The method of item 8035 wherein the agent is a Bruton's tyrosine kinase inhibitor.
8048. The method of item 8035 wherein the agent is a calcineurin inhibitor.
8049. The method of item 8035 wherein the agent is a caspase 3 inhibitor.
8050. The method of item 8035 wherein the agent is a CC chemokine receptor antagonist.
8051. The method of item 8035 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8052. The method of item 8035 wherein the agent is a cathepsin B inhibitor.
8053. The method of item 8035 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8054. The method of item 8035 wherein the agent is a cathepsin L inhibitor.
8055. The method of item 8035 wherein the agent is a CD40 antagonist.
8056. The method of item 8035 wherein the agent is a chemokine receptor agonist. 8057. The method of item 8035 wherein the agent is a chymase inhibitor.
8058. The method of item 8035 wherein the agent is a collagenase antagonist.
8059. The method of item 8035 wherein the agent is a CXCR antagonist.
8060. The method of item 8035 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8061. The method of item 8035 wherein the agent is a cyclooxygenase 1 inhibitor.
8062. The method of item 8035 wherein the agent is a DHFR inhibitor.
8063. The method of item 8035 wherein the agent is a dual integrin inhibitor.
8064. The method of item 8035 wherein the agent is an elastase inhibitor.
8065. The method of item 8035 wherein the agent is an elongation factor-1 alpha inhibitor. 8066. The method of item 8035 wherein the agent is an endothelial growth factor antagonist.
8067. The method of item 8035 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8068. The method of item 8035 wherein the agent is an endotoxin antagonist.
8069. The method of item 8035 wherein the agent is an epothilone and tubulin binder.
8070. The method of item 8035 wherein the agent is an estrogen receptor antagonist.
8071. The method of item 8035 wherein the agent is an FGF inhibitor.
8072. The method of item 8035 wherein the agent is a famexyl transferase inhibitor.
8073. The method of item 8035 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8074. The method of item 8035 wherein the agent is an FLT-3 kinase inhibitor.
8075. The method of item 8035 wherein the agent is an FGF receptor kinase inhibitor.
8076. The method of item 8035 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11 -9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8077. The method of item 8035 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
8078. The method of item 8035 wherein the agent is a histone deacetylase inhibitor.
8079. The method of item 8035 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 8080. The method of item 8035 wherein the agent is an ICAM inhibitor.
8081. The method of item 8035 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
8082. The method of item 8035 wherein the agent is an IL-2 inhibitor.
8083. The method of item 8035 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
8084. The method of item 8035 wherein the agent is an IMPDH (inosine monophosphate). 8085. The method of item 8035 wherein the agent is an integrin antagonist.
8086. The method of item 8035 wherein the agent is an interleukin antagonist.
8087. The method of item 8035 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8088. The method of item 8035 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8089. The method of item 8035 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
8090. The method of item 8035 wherein the agent a JAK3 enzyme inhibitor.
8091. The method of item 8035 wherein the agent is a JNK inhibitor.
8092. The method of item 8035 wherein the agent is a kinase inhibitor.
8093. The method of item 8035 wherein the agent is kinesin antagonist.
8094. The method of item 8035 wherein the agent is a kinesin antagonist.
8095. The method of item 8035 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigei), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8096. The method of item 8035 wherein the agent is an MAP kinase inhibitor.
8097. The method of item 8035 wherein the agent is a matrix metalloproteinase inhibitor.
8098. The method of item 8035 wherein the agent is an MCP-
CCR2 inhibitor.
8099. The method of item 8035 wherein the agent is an mTOR inhibitor.
8100. The method of item 8035 wherein the agent is an mTOR kinase inhibitor.
8101. The method of item 8035 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
8102. The method of item 8035 wherein the agent is an MIF inhibitor.
8103. The method of item 8035 wherein the agent is an MMP inhibitor.
8104. The method of item 8035 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
8105. The method of item 8035 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 8106. The method of item 8035 wherein the agent is a nitric oxide agonist.
8107. The method of item 8035 wherein the agent is an ornithine decarboxylase inhibitor.
8108. The method of item 8035 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8109. The method of item 8035 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8110. The method of item 8035 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
8111. The method of item 8035 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8112. The method of item 8035 wherein the agent is a phosphatase inhibitor.
8113. The method of item 8035 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8114. The method of item 8035 wherein the agent is a PKC inhibitor.
8115. The method of item 8035 wherein the agent is a platelet activating factor antagonist.
8116. The method of item 8035 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
8117. The method of item 8035 wherein the agent is a prolyl hydroxylase inhibitor.
8118. The method of item 8035 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8119. The method of item 8035 wherein the agent is a protein kinase B inhibitor.
8120. The method of item 8035 wherein the agent is a protein kinase C stimulant.
8121. The method of item 8035 wherein the agent is a purine nucleoside analogue.
8122. The method of item 8035 wherein the agent is a purinoreceptor P2X antagonist.
8123. The method of item 8035 wherein the agent is a Raf kinase inhibitor. 8124. The method of item 8035 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8125. The method of item 8035 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
8126. The method of item 8035 wherein the agent is an SDF-1 antagonist.
8127. The method of item 8035 wherein the agent is a sheddase inhibitor.
8128. The method of item 8035 wherein the agent is an SRC inhibitor.
8129. The method of item 8035 wherein the agent is a stromelysin inhibitor.
8130. The method of item 8035 wherein the agent is an Syk kinase inhibitor.
8131. The method of item 8035 wherein the agent is a telomerase inhibitor.
8132. The method of item 8035 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
8133. The method of item 8035 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8134. The method of item 8035 wherein the agent is a Toll receptor inhibitor.
8135. The method of item 8035 wherein the agent is a tubulin antagonist.
8136. The method of item 8035 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8137. The method of item 8035 wherein the agent is a VEGF inhibitor.
8138. The method of item 8035 wherein the agent is a vitamin D receptor agonist.
8139. The method of item 8035 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
8140. The method of item 8035 wherein the agent is AP-23573 (an mTOR inhibitor). 8141. The method of item 8035 wherein the agent is synthadotin (a tubulin antagonist).
8142. The method of item 8035 wherein the agent is S-0885 (a collagenase inhibitor).
8143. The method of item 8035 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
8144. The method of item 8035 wherein the agent is ixabepilone (an epithilone).
8145. The method of item 8035 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
8146. The method of item 8035 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8147. The method of item 8035 wherein the agent is ABT-518 (an angiogenesis inhibitor).
8148. The method of item 8035 wherein the agent is combretastatin (an angiogenesis inhibitor).
8149. The method of item 8035 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8150. The method of item 8035 wherein the agent is SB- 715992 (a kinesin antagonist).
8151. The method of item 8035 wherein the agent is temsirolimus (an mTOR inhibitor).
8152. The method of item 8035 wherein the agent is adalimumab (a TNFα antagonist). 8153. The method of item 8035, wherein the composition comprises a polymer.
8154. The method of item 8035, wherein the composition comprises a polymeric carrier.
8155. The method of item 8035 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
8156. The method of item 8035 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
8157. The method of item 8035 wherein the device has a coating that comprises the anti-scarring agent.
8158. The method of item 8035, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
8159. The method of item 8035, wherein the device has a coating that comprises the agent and directly contacts the sensor.
8160. The method of item 8035, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
8161. The method of item 8035, wherein the device has a coating that comprises the agent and partially covers the sensor.
8162. The method of item 8035, wherein the device has a coating that comprises the agent and completely covers the sensor.
8163. The method of item 8035, wherein the device has a uniform coating.
8164. The method of item 8035, wherein the device has a non- uniform coating. 8165. The method of item 8035, wherein the device has a discontinuous coating.
8166. The method of item 8035, wherein the device has a patterned coating.
8167. The method of item 8035, wherein the device has a coating with a thickness of 100 μm or less.
8168. The method of item 8035, wherein the device has a coating with a thickness of 10 μm or less.
8169. The method of item 8035, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
8170. The method of item 8035, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
8171. The method of item 8035, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
8172. The method of item 8035, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
8173. The method of item 8035, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
8174. The method of item 8035, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 8175. The method of item 8035, wherein the device has a coating, and wherein the coating further comprises a polymer.
8176. The method of item 8035, wherein the device has a first coating having a first composition and a second coating having a second composition.
8177. The method of item 8035, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8178. The method of item 8035, wherein the composition comprises a polymer.
8179. The method of item 8035, wherein the composition comprises a polymeric carrier.
8180. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
8181. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
8182. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
8183. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 8184. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
8185. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
8186. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
8187. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
8188. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
8189. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
8190. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
8191. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
8192. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 8193. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
8194. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
8195. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
8196. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
8197. The method of item 8035, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
8198. The method of item 8035 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
8199. The method of item 8035, wherein the device comprises a lubricious coating.
8200. The method of item 8035 wherein the anti-scarring agent is located within pores or holes of the device.
8201. The method of item 8035 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
8202. The method of item 8035, wherein the device comprises a second pharmaceutically active agent. 8203. The method of item 8035 wherein the device comprises an anti-inflammatory agent.
8204. The method of item 8035 wherein the device comprises an agent that inhibits infection.
8205. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
8206. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
8207. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
8208. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
8209. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
8210. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
8211. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
8212. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
8213. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
8214. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 8215. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
8216. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
8217. The method of item 8035 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
8218. The method of item 8035, further comprising an antithrombotic agent.
8219. The method of item 8035 wherein the device comprises a visualization agent.
8220. The method of item 8035 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
8221. The method of item 8035 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
8222. The method of item 8035 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
8223. The method of item 8035 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 8224. The method of item 8035 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
8225. The method of item 8035 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
8226. The method of item 8035 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
8227. The method of item 8035 wherein the device comprises an echogenic material.
8228. The method of item 8035 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
8229. The method of item 8035 wherein the device is sterile.
8230. The method of item 8035 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
8231. The method of item 8035 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
8232. The method of item 8035 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 8233. The method of item 8035 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
8234. The method of item 8035 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
8235. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
8236. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
8237. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
8238. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
8239. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
8240. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
8241. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 8242. The method of item 8035 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
8243. The method of item 8035 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
8244. The method of item 8035 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
8245. The method of item 8035 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
8246. The method of item 8035 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
8247. The method of item 8035 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
8248. The method of item 8035 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8249. The method of item 8035 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8250. The method of item 8035 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8251. The method of item 8035 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 8252. The method of item 8035 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8253. The method of item 8035 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8254. The method of item 8035 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
8255. The method of item 8035 wherein the combining is performed by spraying the agent or the component onto the sensor.
8256. The method of item 8035 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
8257. The method of item 8035 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
8258. The method of item 8035 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
8259. The method of item 8035 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
8260. The method of item 8035 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
8261. The method of item 8035 wherein the combining is performed by coating the sensor with a substance that absorbs the agent. 8262. The method of item 8035 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
8263. The method of item 8035 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
8264. The method of item 8035 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
8265. The method of item 8035 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
8266. The method of item 8035 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
8267. The method of item 8035 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
8268. The method of item 8035 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
8269. The method of item 8035 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
8270. The method of item 8035 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition. 8271. The method of item 8035 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
8272. The method of item 8035 wherein the combining is performed by impregnating the sensor with the agent or the composition.
8273. The method of item 8035 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
8274. The method of item 8035 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
8275. The method of item 8035 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8276. The method of item 8035 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8277. The method of item 8035 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
8278. The method of item 8035 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8279. The method of item 8035 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 8280. The method of item 8035 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
8281. The method of item 8035 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8282. The method of item 8035 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8283. The method of item 8035 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
8284. The method of item 8035 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8285. The method of item 8035 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
8286. The method of any one of items 8035-8285 wherein the device is adapted for delivering insulin.
8287. The method of any one of items 8035-8285 wherein the device is adapted for delivering a narcotic.
8288. The method of any one of items 8035-8285 wherein the device is adapted for delivering a chemotherapeutic agent.
8289. The method of any one of items 8035-8285 wherein the device is adapted for delivering an anti-arrhythmic drug. 8290. The method of any one of items 8035-8285 wherein the device is adapted for delivering an anti-spasmotic drug.
8291. The method of any one of items 8035-8285 wherein the device is adapted for delivering an anti-spastic agent.
8292. The method of any one of items 8035-8285 wherein the device is adapted for delivering an antibiotic.
8293. The method of any one of items 8035-8285 wherein the device is adapted for delivering a drug only when changes in the host are detected.
8294. The method of any one of items 8035-8285 wherein the device is adapted for delivering a drug as a continuous slow release.
8295. The method of any one of items 8035-8285 wherein the device is adapted for delivering a drug at prescribed dosages in a pulsatile manner.
8296. The method of any one of items 8035-8285 wherein the device is a programmable drug delivery pump.
8297. The method of any one of items 8035-8285 wherein the device is adapted for intraocularly delivering a drug.
8298. The method of any one of items 8035-8285 wherein the device is adapted for intrathecal^ delivering a drug.
8299. The method of any one of items 8035-8285 wherein the device is adapted for intraperitoneal^ delivering a drug.
8300. The method of any one of items 8035-8285 wherein the device is adapted for intra-arterially delivering a drug. 8301. The method of any one of items 8035-8285 wherein the device is adapted for intracardiac delivery of a drug.
8302. The method of any one of items 8035-8285 wherein the device is an implantable osmotic pump.
8303. The method of any one of items 8035-8285 wherein the device is an ocular drug delivery pump.
8304. The method of any one of items 8035-8285 wherein the device is metering system.
8305. The method of any one of items 8035-8285 wherein the device is a peristaltic (roller) pump.
8306. The method of any one of items 8035-8285 wherein the device is an electronically driven pump.
8307. The method of any one of items 8035-8285 wherein the device is an elastomeric pump.
8308. The method of any one of items 8035-8285 wherein the device is a spring contraction pump.
8309. The method of any one of items 8035-8285 wherein the device is a gas-driven pump.
8310. The method of any one of items 8035-8285 wherein the device is a hydraulic pump.
8311. The method of any one of items 8035-8285 wherein the device is a piston-dependent pump.
8312. The method of any one of items 8035-8285 wherein the device is a non-piston-dependent pump. 8313. The method of any one of items 8035-8285 wherein the device is a dispensing chamber.
8314. The method of any one of items 8035-8285 wherein the device is an infusion pump.
8315. The method of any one of items 8035-8285 wherein the device is a passive pump.
8316. A method for making a device comprising: combining an implantable insulin pump and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
8317. The method of item 8316 wherein the agent is an adensosine A2A receptor antagonist.
8318. The method of item 8316 wherein the agent is an AKT inhibitor.
8319. The method of item 8316 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8320. The method of item 8316 wherein the agent is an alpha 4 integrin antagonist.
8321. The method of item 8316 wherein the agent is an alpha 7 nicotinic receptor agonist.
8322. The method of item 8316 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB)1 JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8323. The method of item 8316 wherein the agent is an apoptosis antagonist.
8324. The method of item 8316 wherein the agent is an apoptosis activator. 8325. The method of item 8316 wherein the agent is a beta 1 integrin antagonist.
8326. The method of item 8316 wherein the agent is a beta tubulin inhibitor.
8327. The method of item 8316 wherein the agent is a blocker of enzyme production in Hepatitis C.
8328. The method of item 8316 wherein the agent is a Bruton's tyrosine kinase inhibitor.
8329. The method of item 8316 wherein the agent is a calcineurin inhibitor.
8330. The method of item 8316 wherein the agent is a caspase 3 inhibitor.
8331. The method of item 8316 wherein the agent is a CC chemokine receptor antagonist.
8332. The method of item 8316 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8333. The method of item 8316 wherein the agent is a cathepsin B inhibitor.
8334. The method of item 8316 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8335. The method of item 8316 wherein the agent is a cathepsin L inhibitor. 8336. The method of item 8316 wherein the agent is a CD40 antagonist.
8337. The method of item 8316 wherein the agent is a chemokine receptor agonist. j 8338. The method of item 8316 wherein the agent is a chymase inhibitor.
8339. The method of item 8316 wherein the agent is a collagenase antagonist.
8340. The method of item 8316 wherein the agent is a CXCR antagonist.
8341. The method of item 8316 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8342. The method of item 8316 wherein the agent is a cyclooxygenase 1 inhibitor.
8343. The method of item 8316 wherein the agent is a DHFR inhibitor.
8344. The method of item 8316 wherein the agent is a dual integrin inhibitor. 8345. The method of item 8316 wherein the agent is an elastase inhibitor.
8346. The method of item 8316 wherein the agent is an elongation factor-1 alpha inhibitor.
8347. The method of item 8316 wherein the agent is an endothelial growth factor antagonist.
8348. The method of item 8316 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8349. The method of item 8316 wherein the agent is an endotoxin antagonist.
8350. The method of item 8316 wherein the agent is an epothilone and tubulin binder.
8351. The method of item 8316 wherein the agent is an estrogen receptor antagonist.
8352. The method of item 8316 wherein the agent is an FGF inhibitor.
8353. The method of item 8316 wherein the agent is a farnexyl transferase inhibitor. 8354. The method of item 8316 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8355. The method of item 8316 wherein the agent is an FLT-3 kinase inhibitor.
8356. The method of item 8316 wherein the agent is an FGF receptor kinase inhibitor.
8357. The method of item 8316 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8358. The method of item 8316 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
8359. The method of item 8316 wherein the agent is a histone deacetylase inhibitor.
8360. The method of item 8316 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
8361. The method of item 8316 wherein the agent is an ICAM inhibitor.
8362. The method of item 8316 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
8363. The method of item 8316 wherein the agent is an IL-2 inhibitor.
8364. The method of item 8316 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 8365. The method of item 8316 wherein the agent is an IMPDH (inosine monophosphate).
8366. The method of item 8316 wherein the agent is an integrin antagonist.
8367. The method of item 8316 wherein the agent is an interleukin antagonist.
8368. The method of item 8316 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8369. The method of item 8316 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8370. The method of item 8316 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
8371. The method of item 8316 wherein the agent a JAK3 enzyme inhibitor.
8372. The method of item 8316 wherein the agent is a JNK inhibitor.
8373. The method of item 8316 wherein the agent is a kinase inhibitor.
8374. The method of item 8316 wherein the agent is kinesin antagonist.
8375. The method of item 8316 wherein the agent is a kinesin antagonist.
8376. The method of item 8316 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8377. The method of item 8316 wherein the agent is an MAP kinase inhibitor.
8378. The method of item 8316 wherein the agent is a matrix metalloproteinase inhibitor.
8379. The method of item 8316 wherein the agent is an MCP- CCR2 inhibitor.
8380. The method of item 8316 wherein the agent is an mTOR inhibitor.
8381. The method of item 8316 wherein the agent is an mTOR kinase inhibitor.
8382. The method of item 8316 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Irnotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
8383. The method of item 8316 wherein the agent is an MIF inhibitor.
8384. The method of item 8316 wherein the agent is an MMP inhibitor.
8385. The method of item 8316 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
8386. The method of item 8316 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
8387. The method of item 8316 wherein the agent is a nitric oxide agonist.
8388. The method of item 8316 wherein the agent is an ornithine decarboxylase inhibitor.
8389. The method of item 8316 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8390. The method of item 8316 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8391. The method of item 8316 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
8392. The method of item 8316 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from Eli Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8393. The method of item 8316 wherein the agent is a phosphatase inhibitor.
8394. The method of item 8316 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimiiast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase ill inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8395. The method of item 8316 wherein the agent is a PKC inhibitor.
8396. The method of item 8316 wherein the agent is a platelet activating factor antagonist.
8397. The method of item 8316 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
8398. The method of item 8316 wherein the agent is a prolyl hydroxylase inhibitor.
8399. The method of item 8316 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8400. The method of item 8316 wherein the agent is a protein kinase B inhibitor.
8401. The method of item 8316 wherein the agent is a protein kinase C stimulant.
8402. The method of item 8316 wherein the agent is a purine nucleoside analogue.
8403. The method of item 8316 wherein the agent is a purinoreceptor P2X antagonist. 8404. The method of item 8316 wherein the agent is a Raf kinase inhibitor.
8405. The method of item 8316 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8406. The method of item 8316 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
8407. The method of item 8316 wherein the agent is an SDF-1 antagonist.
8408. The method of item 8316 wherein the agent is a sheddase inhibitor.
8409. The method of item 8316 wherein the agent is an SRC inhibitor.
8410. The method of item 8316 wherein the agent is a stromelysin inhibitor.
8411. The method of item 8316 wherein the agent is an Syk kinase inhibitor.
8412. The method of item 8316 wherein the agent is a telomerase inhibitor.
8413. The method of item 8316 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No.
53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (In2Gen), mannose-6-phosphate (BTG)1 TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 8414. The method of item 8316 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPFM 32294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8415. The method of item 8316 wherein the agent is a Toll receptor inhibitor.
8416. The method of item 8316 wherein the agent is a tubulin antagonist.
8417. The method of item 8316 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8418. The method of item 8316 wherein the agent is a VEGF inhibitor.
8419. The method of item 8316 wherein the agent is a vitamin D receptor agonist.
8420. The method of item 8316 wherein the agent is ZD-6474 (an angiogenesis inhibitor). 8421. The method of item 8316 wherein the agent is AP-23573 (an mTOR inhibitor).
8422. The method of item 8316 wherein the agent is synthadotin (a tubulin antagonist).
8423. The method of item 8316 wherein the agent is S-0885 (a collagenase inhibitor).
8424. The method of item 8316 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
8425. The method of item 8316 wherein the agent is ixabepilone (an epithilone).
8426. The method of item 8316 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
8427. The method of item 8316 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8428. The method of item 8316 wherein the agent is ABT-518
(an angiogenesis inhibitor).
8429. The method of item 8316 wherein the agent is combretastatin (an angiogenesis inhibitor).
8430. The method of item 8316 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8431. The method of item 8316 wherein the agent is SB- 715992 (a kinesin antagonist).
8432. The method of item 8316 wherein the agent is temsirolimus (an mTOR inhibitor). 8433. The method of item 8316 wherein the agent is adalimumab (a TNFα antagonist).
8434. The method of item 8316, wherein the composition comprises a polymer.
8435. The method of item 8316, wherein the composition comprises a polymeric carrier.
8436. The method of item 8316 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
8437. The method of item 8316 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
8438. The method of item 8316 wherein the device has a coating that comprises the anti-scarring agent.
8439. The method of item 8316, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
8440. The method of item 8316, wherein the device has a coating that comprises the agent and directly contacts the sensor.
8441. The method of item 8316, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
8442. The method of item 8316, wherein the device has a coating that comprises the agent and partially covers the sensor.
8443. The method of item 8316, wherein the device has a coating that comprises the agent and completely covers the sensor.
8444. The method of item 8316, wherein the device has a uniform coating. 8445. The method of item 8316, wherein the device has a nonuniform coating.
8446. The method of item 8316, wherein the device has a discontinuous coating.
8447. The method of item 8316, wherein the device has a patterned coating.
8448. The method of item 8316, wherein the device has a coating with a thickness of 100 μm or less.
8449. The method of item 8316, wherein the device has a coating with a thickness of 10 μm or less.
8450. The method of item 8316, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
8451. The method of item 8316, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
8452. The method of item 8316, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
8453. The method of item 8316, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
8454. The method of item 8316, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 8455. The method of item 8316, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
8456. The method of item 8316, wherein the device has a coating, and wherein the coating further comprises a polymer.
8457. The method of item 8316, wherein the device has a first coating having a first composition and a second coating having a second composition.
8458. The method of item 8316, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8459. The method of item 8316, wherein the composition comprises a polymer.
8460. The method of item 8316, wherein the composition comprises a polymeric carrier.
8461. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
8462. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
8463. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer. 8464. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
8465. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
8466. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
8467. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
8468. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
8469. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
8470. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
8471. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
8472. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel. 8473. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
8474. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
8475. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
8476. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
8477. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
8478. The method of item 8316, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a po!y(ethylene glycol) polymer.
8479. The method of item 8316 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
8480. The method of item 8316, wherein the device comprises a lubricious coating.
8481. The method of item 8316 wherein the anti-scarring agent is located within pores or holes of the device. 8482. The method of item 8316 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
8483. The method of item 8316, wherein the device comprises a second pharmaceutically active agent.
8484. The method of item 8316 wherein the device comprises an anti-inflammatory agent.
8485. The method of item 8316 wherein the device comprises an agent that inhibits infection.
8486. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
8487. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
8488. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
8489. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
8490. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
8491. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
8492. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
8493. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin. 8494. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
8495. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
8496. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
8497. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
8498. The method of item 8316 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
8499. The method of item 8316, further comprising an antithrombotic agent.
8500. The method of item 8316 wherein the device comprises a visualization agent.
8501. The method of item 8316 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
8502. The method of item 8316 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
8503. The method of item 8316 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material. 8504. The method of item 8316 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
8505. The method of item 8316 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
8506. The method of item 8316 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
8507. The method of item 8316 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
8508. The method of item 8316 wherein the device comprises an echogenic material.
8509. The method of item 8316 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
8510. The method of item 8316 wherein the device is sterile.
8511. The method of item 8316 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
8512. The method of item 8316 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue. 8513. The method of item 8316 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
8514. The method of item 8316 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
8515. The method of item 8316 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
8516. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
8517. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
8518. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
8519. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
8520. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
8521. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
8522. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
8523. The method of item 8316 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
8524. The method of item 8316 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
8525. The method of item 8316 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
8526. The method of item 8316 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
8527. The method of item 8316 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
8528. The method of item 8316 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
8529. The method of item 8316 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8530. The method of item 8316 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8531. The method of item 8316 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 8532. The method of item 8316 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8533. The method of item 8316 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8534. The method of item 8316 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8535. The method of item 8316 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
8536. The method of item 8316 wherein the combining is performed by spraying the agent or the component onto the sensor.
8537. The method of item 8316 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
8538. The method of item 8316 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
8539. The method of item 8316 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
8540. The method of item 8316 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor. 8541. The method of item 8316 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
8542. The method of item 8316 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
8543. The method of item 8316 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
8544. The method of item 8316 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
8545. The method of item 8316 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
8546. The method of item 8316 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
8547. The method of item 8316 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
8548. The method of item 8316 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
8549. The method of item 8316 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition. 8550. The method of item 8316 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
8551. The method of item 8316 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
8552. The method of item 8316 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
8553. The method of item 8316 wherein the combining is performed by impregnating the sensor with the agent or the composition.
8554. The method of item 8316 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
8555. The method of item 8316 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
8556. The method of item 8316 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8557. The method of item 8316 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8558. The method of item 8316 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor. 8559. The method of item 8316 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8560. The method of item 8316 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent'that will dissolve the sensor.
8561. The method of item 8316 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
8562. The method of item 8316 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8563. The method of item 8316 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8564. The method of item 8316 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
8565. The method of item 8316 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8566. The method of item 8316 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
8567. The method of any one of items 8316-8566 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense mediation through the catheter.
8568. A method for making a device comprising: combining an intrathecal drug delivery pump and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
8569. The method of item 8568 wherein the agent is an adensosine A2A receptor antagonist.
8570. The method of item 8568 wherein the agent is an AKT inhibitor.
8571. The method of item 8568 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8572. The method of item 8568 wherein the agent is an alpha 4 integrin antagonist.
8573. The method of item 8568 wherein the agent is an alpha 7 nicotinic receptor agonist.
8574. The method of item 8568 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore
Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attention), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT- 116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8575. The method of item 8568 wherein the agent is an apoptosis antagonist.
8576. The method of item 8568 wherein the agent is an apoptosis activator.
8577. The method of item 8568 wherein the agent is a beta 1 integrin antagonist.
8578. The method of item 8568 wherein the agent is a beta tubulin inhibitor. 8579. The method of item 8568 wherein the agent is a blocker of enzyme production in Hepatitis C.
8580. The method of item 8568 wherein the agent is a Bruton's tyrosine kinase inhibitor.
8581. The method of item 8568 wherein the agent is a calcineurin inhibitor.
8582. The method of item 8568 wherein the agent is a caspase 3 inhibitor.
8583. The method of item 8568 wherein the agent is a CC chemokine receptor antagonist.
8584. The method of item 8568 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotln, KRX-0403, and an analogue or derivative thereof.
8585. The method of item 8568 wherein the agent is a cathepsin B inhibitor.
8586. The method of item 8568 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8587. The method of item 8568 wherein the agent is a cathepsin L inhibitor.
8588. The method of item 8568 wherein the agent is a CD40 antagonist.
8589. The method of item 8568 wherein the agent is a chemokine receptor agonist. 8590. The method of item 8568 wherein the agent is a chymase inhibitor.
8591. The method of item 8568 wherein the agent is a collagenase antagonist.
8592. The method of item 8568 wherein the agent is a CXCR antagonist.
8593. The method of item 8568 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8594. The method of item 8568 wherein the agent is a cyclooxygenase 1 inhibitor.
8595. The method of item 8568 wherein the agent is a DHFR inhibitor.
8596. The method of item 8568 wherein the agent is a dual integrin inhibitor.
8597. The method of item 8568 wherein the agent is an elastase inhibitor.
8598. The method of item 8568 wherein the agent is an elongation factor-1 alpha inhibitor. 8599. The method of item 8568 wherein the agent is an endothelial growth factor antagonist.
8600. The method of item 8568 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8601. The method of item 8568 wherein the agent is an endotoxin antagonist.
8602. The method of item 8568 wherein the agent is an epothilone and tubulin binder.
8603. The method of item 8568 wherein the agent is an estrogen receptor antagonist.
8604. The method of item 8568 wherein the agent is an FGF inhibitor.
8605. The method of item 8568 wherein the agent is a famexyl transferase inhibitor.
8606. The method of item 8568 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8607. The method of item 8568 wherein the agent is an FLT-3 kinase inhibitor.
8608. The method of item 8568 wherein the agent is an FGF receptor kinase inhibitor.
8609. The method of item 8568 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8610. The method of item 8568 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
8611. The method of item 8568 wherein the agent is a histone deacetylase inhibitor.
8612. The method of item 8568 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 8613. The method of item 8568 wherein the agent is an ICAM inhibitor.
8614. The method of item 8568 wherein the agent is an IL1 ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
8615. The method of item 8568 wherein the agent is an IL-2 inhibitor.
8616. The method of item 8568 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
8617. The method of item 8568 wherein the agent is an IMPDH (inosine monophosphate). 8618. The method of item 8568 wherein the agent is an integrin antagonist.
8619. The method of item 8568 wherein the agent is an interleukin antagonist.
8620. The method of item 8568 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8621. The method of item 8568 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8622. The method of item 8568 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
8623. The method of item 8568 wherein the agent a JAK3 enzyme inhibitor.
8624. The method of item 8568 wherein the agent is a JNK inhibitor.
8625. The method of item 8568 wherein the agent is a kinase inhibitor.
8626. The method of item 8568 wherein the agent is kinesin antagonist.
8627. The method of item 8568 wherein the agent is a kinesin antagonist.
8628. The method of item 8568 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, Iymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8629. The method of item 8568 wherein the agent is an MAP kinase inhibitor.
8630. The method of item 8568 wherein the agent is a matrix metalloproteinase inhibitor.
8631. The method of item 8568 wherein the agent is an MCP-
CCR2 inhibitor.
8632. The method of item 8568 wherein the agent is an mTOR inhibitor.
8633. The method of item 8568 wherein the agent is an mTOR kinase inhibitor.
8634. The method of item 8568 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
8635. The method of item 8568 wherein the agent is an MIF inhibitor.
8636. The method of item 8568 wherein the agent is an MMP inhibitor.
8637. The method of item 8568 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
8638. The method of item 8568 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 8639. The method of item 8568 wherein the agent is a nitric oxide agonist.
8640. The method of item 8568 wherein the agent is an ornithine decarboxylase inhibitor.
8641. The method of item 8568 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8642. The method of item 8568 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8643. The method of item 8568 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
8644. The method of item 8568 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8645. The method of item 8568 wherein the agent is a phosphatase inhibitor.
8646. The method of item 8568 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8647. The method of item 8568 wherein the agent is a PKC inhibitor.
8648. The method of item 8568 wherein the agent is a platelet activating factor antagonist.
8649. The method of item 8568 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
8650. The method of item 8568 wherein the agent is a prolyl hydroxylase inhibitor.
8651. The method of item 8568 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8652. The method of item 8568 wherein the agent is a protein kinase B inhibitor.
8653. The method of item 8568 wherein the agent is a protein kinase C stimulant.
8654. The method of item 8568 wherein the agent is a purine nucleoside analogue.
8655. The method of item 8568 wherein the agent is a purinoreceptor P2X antagonist.
8656. The method of item 8568 wherein the agent is a Raf kinase inhibitor. 8657. The method of item 8568 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8658. The method of item 8568 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
8659. The method of item 8568 wherein the agent is an SDF-1 antagonist.
8660. The method of item 8568 wherein the agent is a sheddase inhibitor.
8661. The method of item 8568 wherein the agent is an SRC inhibitor.
8662. The method of item 8568 wherein the agent is a stromelysin inhibitor.
8663. The method of item 8568 wherein the agent is an Syk kinase inhibitor.
8664. The method of item 8568 wherein the agent is a telomerase inhibitor.
8665. The method of item 8568 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
8666. The method of item 8568 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8667. The method of item 8568 wherein the agent is a Toll receptor inhibitor.
8668. The method of item 8568 wherein the agent is a tubulin antagonist.
8669. The method of item 8568 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8670. The method of item 8568 wherein the agent is a VEGF inhibitor.
8671. The method of item 8568 wherein the agent is a vitamin D receptor agonist.
8672. The method of item 8568 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
8673. The method of item 8568 wherein the agent is AP-23573 (an mTOR inhibitor). 8674. The method of item 8568 wherein the agent is synthadotin (a tubulin antagonist).
8675. The method of item 8568 wherein the agent is S-0885 (a collagenase inhibitor).
8676. The method of item 8568 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
8677. The method of item 8568 wherein the agent is ixabepilone (an epithilone).
8678. The method of item 8568 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
8679. The method of item 8568 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8680. The method of item 8568 wherein the agent is ABT-518 (an angiogenesis inhibitor).
8681. The method of item 8568 wherein the agent is combretastatin (an angiogenesis inhibitor).
8682. The method of item 8568 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8683. The method of item 8568 wherein the agent is SB- 715992 (a kinesin antagonist).
8684. The method of item 8568 wherein the agent is temsirolimus (an mTOR inhibitor).
8685. The method of item 8568 wherein the agent is adalimumab (a TNFα antagonist). 8686. The method of item 8568, wherein the composition comprises a polymer.
8687. The method of item 8568, wherein the composition comprises a polymeric carrier.
8688. The method of item 8568 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
8689. The method of item 8568 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
8690. The method of item 8568 wherein the device has a coating that comprises the anti-scarring agent.
8691. The method of item 8568, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
8692. The method of item 8568, wherein the device has a coating that comprises the agent and directly contacts the sensor.
8693. The method of item 8568, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
8694. The method of item 8568, wherein the device has a coating that comprises the agent and partially covers the sensor.
8695. The method of item 8568, wherein the device has a coating that comprises the agent and completely covers the sensor.
8696. The method of item 8568, wherein the device has a uniform coating.
8697. The method of item 8568, wherein the device has a non- uniform coating. 8698. The method of item 8568, wherein the device has a discontinuous coating.
8699. The method of item 8568, wherein the device has a patterned coating.
8700. The method of item 8568, wherein the device has a coating with a thickness of 100 μm or less.
8701. The method of item 8568, wherein the device has a coating with a thickness of 10 μm or less.
8702. The method of item 8568, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
8703. The method of item 8568, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
8704. The method of item 8568, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1% by weight.
8705. The method of item 8568, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
8706. The method of item 8568, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
8707. The method of item 8568, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 8708. The method of item 8568, wherein the device has a coating, and wherein the coating further comprises a polymer.
8709. The method of item 8568, wherein the device has a first coating having a first composition and a second coating having a second composition.
8710. The method of item 8568, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8711. The method of item 8568, wherein the composition comprises a polymer.
8712. The method of item 8568, wherein the composition comprises a polymeric carrier.
8713. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
8714. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
8715. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
8716. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 8717. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
8718. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
8719. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
8720. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
8721. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
8722. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
8723. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
8724. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
8725. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 8726. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
8727. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
8728. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
8729. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
8730. The method of item 8568, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
8731. The method of item 8568 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
8732. The method of item 8568, wherein the device comprises a lubricious coating.
8733. The method of item 8568 wherein the anti-scarring agent is located within pores or holes of the device.
8734. The method of item 8568 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
8735. The method of item 8568, wherein the device comprises a second pharmaceutically active agent. 8736. The method of item 8568 wherein the device comprises an anti-inflammatory agent.
8737. The method of item 8568 wherein the device comprises an agent that inhibits infection.
8738. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
8739. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
8740. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
8741. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
8742. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
8743. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
8744. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
8745. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
8746. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
8747. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 8748. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
8749. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
8750. The method of item 8568 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
8751. The method of item 8568, further comprising an antithrombotic agent.
8752. The method of item 8568 wherein the device comprises a visualization agent.
8753. The method of item 8568 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
8754. The method of item 8568 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
8755. The method of item 8568 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
8756. The method of item 8568 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 8757. The method of item 8568 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
8758. The method of item 8568 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. .
8759. The method of item 8568 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
8760. The method of item 8568 wherein the device comprises an echogenic material.
8761. The method of item 8568 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
8762. The method of item 8568 wherein the device is sterile.
8763. The method of item 8568 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
8764. The method of item 8568 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
8765. The method of item 8568 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 8766. The method of item 8568 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
8767. The method of item 8568 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
8768. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
8769. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
8770. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
8771. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
8772. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
8773. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
8774. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 8775. The method of item 8568 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
8776. The method of item 8568 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
8777. The method of item 8568 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
8778. The method of item 8568 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
8779. The method of item 8568 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
8780. The method of item 8568 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
8781. The method of item 8568 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8782. The method of item 8568 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8783. The method of item 8568 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8784. The method of item 8568 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 8785. The method of item 8568 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8786. The method of item 8568 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
8787. The method of item 8568 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
8788. The method of item 8568 wherein the combining is performed by spraying the agent or the component onto the sensor.
8789. The method of item 8568 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
8790. The method of item 8568 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
8791. The method of item 8568 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
8792. The method of item 8568 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
8793. The method of item 8568 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
8794. The method of item 8568 wherein the combining is performed by coating the sensor with a substance that absorbs the agent. 8795. The method of item 8568 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
8796. The method of item 8568 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
8797. The method of item 8568 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
8798. The method of item 8568 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
8799. The method of item 8568 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
8800. The method of item 8568 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
8801. The method of item 8568 wherein the combining is performed by covering a portion of the sensor with an eiectrospun fabric that contains the agent or the composition.
8802. The method of item 8568 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
8803. The method of item 8568 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition. 8804. The method of item 8568 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
8805. The method of item 8568 wherein the combining is performed by impregnating the sensor with the agent or the composition.
8806. The method of item 8568 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
8807. The method of item 8568 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
8808. The method of item 8568 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8809. The method of item 8568 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8810. The method of item 8568 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
8811. The method of item 8568 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8812. The method of item 8568 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 8813. The method of item 8568 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
8814. The method of item 8568 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
8815. The method of item 8568 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
8816. The method of item 8568 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
8817. The method of item 8568 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
8818. The method of item 8568 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
8819. The method of any one of items 8568-8818 wherein the device is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord.
8820. The method of any one of items 8568-8818 wherein the device is adapted for delivering a drug to the brain.
8821. The method of any one of items 8568-8818 wherein the device is adapted for intrathecal delivering baclofen. 8822. The method of any one of items 8568-8818 wherein the device further comprises an intraspinal catheter.
8823. The method of any one of items 8568-8818 further comprising a second intrathecal drug delivery pump.
8824. The method of any one of items 8568-8818 wherein the device further comprises a catheter and an electrode.
8825. A method for making a device comprising: combining an implantable drug delivery pump for chemotherapy and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
8826. The method of item 8825 wherein the agent is an adensosine A2A receptor antagonist.
8827. The method of item 8825 wherein the agent is an AKT inhibitor.
8828. The method of item 8825 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8829. The method of item 8825 wherein the agent is an alpha 4 integrin antagonist.
8830. The method of item 8825 wherein the agent is an alpha 7 nicotinic receptor agonist.
8831. The method of item 8825 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), K1N-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH)1 WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8832. The method of item 8825 wherein the agent is an apoptosis antagonist.
8833. The method of item 8825 wherein the agent is an apoptosis activator. 8834. The method of item 8825 wherein the agent is a beta 1 integrin antagonist.
8835. The method of item 8825 wherein the agent is a beta tubulin inhibitor.
8836. The method of item 8825 wherein the agent is a blocker of enzyme production in Hepatitis C.
8837. The method of item 8825 wherein the agent is a Bruton's tyrosine kinase inhibitor.
8838. The method of item 8825 wherein the agent is a calcineurin inhibitor.
8839. The method of item 8825 wherein the agent is a caspase 3 inhibitor.
8840. The method of item 8825 wherein the agent is a CC chemokine receptor antagonist.
8841. The method of item 8825 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8842. The method of item 8825 wherein the agent is a cathepsin B inhibitor.
8843. The method of item 8825 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8844. The method of item 8825 wherein the agent is a cathepsin L inhibitor. 8845. The method of item 8825 wherein the agent is a CD40 antagonist.
8846. The method of item 8825 wherein the agent is a chemokine receptor agonist.
8847. The method of item 8825 wherein the agent is a chymase inhibitor.
8848. The method of item 8825 wherein the agent is a collagenase antagonist.
8849. The method of item 8825 wherein the agent is a CXCR antagonist.
8850. The method of item 8825 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (Eli Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8851. The method of item 8825 wherein the agent is a cyclooxygenase 1 inhibitor.
8852. The method of item 8825 wherein the agent is a DHFR inhibitor.
8853. The method of item 8825 wherein the agent is a dual integrin inhibitor. 8854. The method of item 8825 wherein the agent is an elastase inhibitor.
8855. The method of item 8825 wherein the agent is an elongation factor-1 alpha inhibitor.
8856. The method of item 8825 wherein the agent is an endothelial growth factor antagonist.
8857. The method of item 8825 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8858. The method of item 8825 wherein the agent is an endotoxin antagonist.
8859. The method of item 8825 wherein the agent is an epothilone and tubulin binder.
8860. The method of item 8825 wherein the agent is an estrogen receptor antagonist.
8861. The method of item 8825 wherein the agent is an FGF inhibitor.
8862. The method of item 8825 wherein the agent is a farnexyl transferase inhibitor. 8863. The method of item 8825 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8864. The method of item 8825 wherein the agent is an FLT-3 kinase inhibitor.
8865. The method of item 8825 wherein the agent is an FGF receptor kinase inhibitor.
8866. The method of item 8825 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8867. The method of item 8825 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
8868. The method of item 8825 wherein the agent is a histone deacetylase inhibitor.
8869. The method of item 8825 wherein the agent is an
HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
8870. The method of item 8825 wherein the agent is an ICAM inhibitor.
8871. The method of item 8825 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
8872. The method of item 8825 wherein the agent is an IL-2 inhibitor.
8873. The method of item 8825 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 8874. The method of item 8825 wherein the agent is an IMPDH (inosine monophosphate).
8875. The method of item 8825 wherein the agent is an integrin antagonist.
8876. The method of item 8825 wherein the agent is an interleukin antagonist.
8877. The method of item 8825 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8878. The method of item 8825 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8879. The method of item 8825 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
8880. The method of item 8825 wherein the agent a JAK3 enzyme inhibitor.
8881. The method of item 8825 wherein the agent is a JNK inhibitor.
8882. The method of item 8825 wherein the agent is a kinase inhibitor.
8883. The method of item 8825 wherein the agent is kinesin antagonist.
8884. The method of item 8825 wherein the agent is a kinesin antagonist.
8885. The method of item 8825 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8886. The method of item 8825 wherein the agent is an MAP kinase inhibitor.
8887. The method of item 8825 wherein the agent is a matrix metalloproteinase inhibitor.
8888. The method of item 8825 wherein the agent is an MCP- CCR2 inhibitor.
8889. The method of item 8825 wherein the agent is an mTOR inhibitor.
8890. The method of item 8825 wherein the agent is an mTOR kinase inhibitor.
8891. The method of item 8825 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
8892. The method of item 8825 wherein the agent is an MIF inhibitor.
8893. The method of item 8825 wherein the agent is an MMP inhibitor.
8894. The method of item 8825 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
8895. The method of item 8825 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
8896. The method of item 8825 wherein the agent is a nitric oxide agonist.
8897. The method of item 8825 wherein the agent is an ornithine decarboxylase inhibitor.
8898. The method of item 8825 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8899. The method of item 8825 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8900. The method of item 8825 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
8901. The method of item 8825 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL7 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8902. The method of item 8825 wherein the agent is a phosphatase inhibitor.
8903. The method of item 8825 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8904. The method of item 8825 wherein the agent is a PKC inhibitor.
8905. The method of item 8825 wherein the agent is a platelet activating factor antagonist.
8906. The method of item 8825 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
8907. The method of item 8825 wherein the agent is a prolyl hydroxylase inhibitor.
8908. The method of item 8825 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8909. The method of item 8825 wherein the agent is a protein kinase B inhibitor.
8910. The method of item 8825 wherein the agent is a protein kinase C stimulant.
8911. The method of item 8825 wherein the agent is a purine nucleoside analogue.
8912. The method of item 8825 wherein the agent is a purinoreceptor P2X antagonist. 8913. The method of item 8825 wherein the agent is a Raf kinase inhibitor.
8914. The method of item 8825 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8915. The method of item 8825 wherein the agent is a hbonucleoside triphosphate reductase inhibitor.
8916. The method of item 8825 wherein the agent is an SDF-1 antagonist.
8917. The method of item 8825 wherein the agent is a sheddase inhibitor.
8918. The method of item 8825 wherein the agent is an SRC inhibitor.
8919. The method of item 8825 wherein the agent is a stromelysin inhibitor.
8920. The method of item 8825 wherein the agent is an Syk kinase inhibitor.
8921. The method of item 8825 wherein the agent is a telomerase inhibitor.
8922. The method of item 8825 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No.
53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 8923. The method of item 8825 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8924. The method of item 8825 wherein the agent is a Toll receptor inhibitor.
8925. The method of item 8825 wherein the agent is a tubulin antagonist.
8926. The method of item 8825 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8927. The method of item 8825 wherein the agent is a VEGF inhibitor.
8928. The method of item 8825 wherein the agent is a vitamin D receptor agonist.
8929. The method of item 8825 wherein the agent is ZD-6474 (an angiogenesis inhibitor). 8930. The method of item 8825 wherein the agent is AP-23573 (an mTOR inhibitor).
8931. The method of item 8825 wherein the agent is synthadotin (a tubulin antagonist).
8932. The method of item 8825 wherein the agent is S-0885 (a collagenase inhibitor).
8933. The method of item 8825 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
8934. The method of item 8825 wherein the agent is ixabepilone (an epithilone).
8935. The method of item 8825 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
8936. The method of item 8825 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8937. The method of item 8825 wherein the agent is ABT-518
(an angiogenesis inhibitor).
8938. The method of item 8825 wherein the agent is combretastatin (an angiogenesis inhibitor).
8939. The method of item 8825 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8940. The method of item 8825 wherein the agent is SB- 715992 (a kinesin antagonist).
8941. The method of item 8825 wherein the agent is temsirolimus (an mTOR inhibitor). 8942. The method of item 8825 wherein the agent is adalimumab (a TNFα antagonist).
8943. The method of item 8825, wherein the composition comprises a polymer.
8944. The method of item 8825, wherein the composition comprises a polymeric carrier.
8945. The method of item 8825 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
8946. The method of item 8825 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
8947. The method of item 8825 wherein the device has a coating that comprises the anti-scarring agent.
8948. The method of item 8825, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
8949. The method of item 8825, wherein the device has a coating that comprises the agent and directly contacts the sensor.
8950. The method of item 8825, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
8951. The method of item 8825, wherein the device has a coating that comprises the agent and partially covers the sensor.
8952. The method of item 8825, wherein the device has a coating that comprises the agent and completely covers the sensor.
8953. The method of item 8825, wherein the device has a uniform coating. 8954. The method of item 8825, wherein the device has a nonuniform coating.
8955. The method of item 8825, wherein the device has a discontinuous coating.
8956. The method of item 8825, wherein the device has a patterned coating.
8957. The method of item 8825, wherein the device has a coating with a thickness of 100 μm or less.
8958. The method of item 8825, wherein the device has a coating with a thickness of 10 μm or less.
8959. The method of item 8825, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
8960. The method of item 8825, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
8961. The method of item 8825, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
8962. The method of item 8825, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
8963. The method of item 8825, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 8964. The method of item 8825, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
8965. The method of item 8825, wherein the device has a coating, and wherein the coating further comprises a polymer.
8966. The method of item 8825, wherein the device has a first coating having a first composition and a second coating having a second composition.
8967. The method of item 8825, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8968. The method of item 8825, wherein the composition comprises a polymer.
8969. The method of item 8825, wherein the composition comprises a polymeric carrier.
8970. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
8971. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
8972. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer. 8973. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
8974. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
8975. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
8976. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
8977. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
8978. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
8979. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
8980. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
8981. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel. 8982. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
8983. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
8984. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
8985. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
8986. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
8987. The method of item 8825, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
8988. The method of item 8825 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
8989. The method of item 8825, wherein the device comprises a lubricious coating.
8990. The method of item 8825 wherein the anti-scarring agent is located within pores or holes of the device. 8991. The method of item 8825 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
8992. The method of item 8825, wherein the device comprises a second pharmaceutically active agent.
8993. The method of item 8825 wherein the device comprises an anti-inflammatory agent.
8994. The method of item 8825 wherein the device comprises an agent that inhibits infection.
8995. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
8996. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
8997. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
8998. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
8999. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
9000. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
9001. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
9002. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin. 9003. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
9004. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
9005. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
9006. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
9007. The method of item 8825 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
9008. The method of item 8825, further comprising an antithrombotic agent.
9009. The method of item 8825 wherein the device comprises a visualization agent.
9010. The method of item 8825 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
9011. The method of item 8825 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
9012. The method of item 8825 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material. 9013. The method of item 8825 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
9014. The method of item 8825 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
9015. The method of item 8825 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
9016. The method of item 8825 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
9017. The method of item 8825 wherein the device comprises an echogenic material.
9018. The method of item 8825 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
9019. The method of item 8825 wherein the device is sterile.
9020. The method of item 8825 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
9021. The method of item 8825 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue. is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
9023. The method of item 8825 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
9024. The method of item 8825 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
9025. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
9026. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
9027. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
9028. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
9029. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
9030. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
9031. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
9032. The method of item 8825 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
9033. The method of item 8825 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
9034. The method of item 8825 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
9035. The method of item 8825 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
9036. The method of item 8825 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
9037. The method of item 8825 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
9038. The method of item 8825 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9039. The method of item 8825 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9040. The method of item 8825 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 9041. The method of item 8825 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9042. The method of item 8825 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9043. The method of item 8825 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9044. The method of item 8825 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
9045. The method of item 8825 wherein the combining is performed by spraying the agent or the component onto the sensor.
9046. The method of item 8825 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
9047. The method of item 8825 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
9048. The method of item 8825 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
9049. The method of item 8825 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor. 9050. The method of item 8825 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
9051. The method of item 8825 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
9052. The method of item 8825 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
9053. The method of item 8825 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
9054. The method of item 8825 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
9055. The method of item 8825 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
9056. The method of item 8825 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
9057. The method of item 8825 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
9058. The method of item 8825 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition. 9059. The method of item 8825 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
9060. The method of item 8825 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
9061. The method of item 8825 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
9062. The method of item 8825 wherein the combining is performed by impregnating the sensor with the agent or the composition.
9063. The method of item 8825 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
9064. The method of item 8825 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
9065. The method of item 8825 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
9066. The method of item 8825 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
9067. The method of item 8825 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor. 9068. The method of item 8825 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
9069. The method of item 8825 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
9070. The method of item 8825 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
9071. The method of item 8825 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
9072. The method of item 8825 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
9073. The method of item 8825 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
9074. The method of item 8825 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
9075. The method of item 8825 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
9076. The method of any one of items 8825-9075 wherein the device is adapted for delivering 2'-deoxy 5-fluorouridine. 9077. The method of any one of items 8825-9075 wherein the host has a solid tumor, and the device is adapted for infusing a chemotherapeutic agent to the solid tumor.
9078. The method of any one of items 8825-9075 wherein the host has a tumor, and the device is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
9079. The method of any one of items 8825-9075 wherein the host has a hepatic tumor, and the device is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host.
9080. A method for making a device comprising: combining a drug delivery pump for treating heart disease and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
9081. The method of item 9080 wherein the agent is an adensosine A2A receptor antagonist.
9082. The method of item 9080 wherein the agent is an AKT inhibitor.
9083. The method of item 9080 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects
No. 5754 (Merck KgaA).
9084. The method of item 9080 wherein the agent is an alpha 4 integrin antagonist.
9085. The method of item 9080 wherein the agent is an alpha 7 nicotinic receptor agonist. 9086. The method of item 9080 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 9087. The method of item 9080 wherein the agent is an apoptosis antagonist.
9088. The method of item 9080 wherein the agent is an apoptosis activator.
9089. The method of item 9080 wherein the agent is a beta 1 integrin antagonist.
9090. The method of item 9080 wherein the agent is a beta tubulin inhibitor.
9091. The method of item 9080 wherein the agent is a blocker of enzyme production in Hepatitis C.
9092. The method of item 9080 wherein the agent is a Bruton's tyrosine kinase inhibitor.
9093. The method of item 9080 wherein the agent is a calcineurin inhibitor.
9094. The method of item 9080 wherein the agent is a caspase
3 inhibitor.
9095. The method of item 9080 wherein the agent is a CC chemokine receptor antagonist.
9096. The method of item 9080 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin,
KRX-0403, and an analogue or derivative thereof.
9097. The method of item 9080 wherein the agent is a cathepsin B inhibitor.
9098. The method of item 9080 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
9099. The method of item 9080 wherein the agent is a cathepsin L inhibitor.
9100. The method of item 9080 wherein the agent is a CD40 antagonist.
9101. The method of item 9080 wherein the agent is a chemokine receptor agonist.
9102. The method of item 9080 wherein the agent is a chymase inhibitor.
9103. The method of item 9080 wherein the agent is a collagenase antagonist.
9104. The method of item 9080 wherein the agent is a CXCR antagonist.
9105. The method of item 9080 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
9106. The method of item 9080 wherein the agent is a cyclooxygenase 1 inhibitor. 9107. The method of item 9080 wherein the agent is a DHFR inhibitor.
9108. The method of item 9080 wherein the agent is a dual integrin inhibitor.
9109. The method of item 9080 wherein the agent is an elastase inhibitor.
9110. The method of item 9080 wherein the agent is an elongation factor-1 alpha inhibitor.
9111. The method of item 9080 wherein the agent is an endothelial growth factor antagonist.
9112. The method of item 9080 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tϊe-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
9113. The method of item 9080 wherein the agent is an endotoxin antagonist.
9114. The method of item 9080 wherein the agent is an epothilone and tubulin binder.
9115. The method of item 9080 wherein the agent is an estrogen receptor antagonist. 9116. The method of item 9080 wherein the agent is an FGF inhibitor.
9117. The method of item 9080 wherein the agent is a farnexyl transferase inhibitor.
9118. The method of item 9080 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9119. The method of item 9080 wherein the agent is an FLT-3 kinase inhibitor.
9120. The method of item 9080 wherein the agent is an FGF receptor kinase inhibitor.
9121. The method of item 9080 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9122. The method of item 9080 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof. 9123. The method of item 9080 wherein the agent is a histone deacetylase inhibitor.
9124. The method of item 9080 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9125. The method of item 9080 wherein the agent is an ICAM inhibitor.
9126. The method of item 9080 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9127. The method of item 9080 wherein the agent is an IL-2 inhibitor.
9128. The method of item 9080 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus
Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9129. The method of item 9080 wherein the agent is an IMPDH (inosine monophosphate).
9130. The method of item 9080 wherein the agent is an integrin antagonist.
9131. The method of item 9080 wherein the agent is an interleukin antagonist.
9132. The method of item 9080 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
9133. The method of item 9080 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9134. The method of item 9080 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
9135. The method of item 9080 wherein the agent a JAK3 enzyme inhibitor.
9136. The method of item 9080 wherein the agent is a JNK inhibitor.
9137. The method of item 9080 wherein the agent is a kinase inhibitor.
9138. The method of item 9080 wherein the agent is kinesin antagonist. 9139. The method of item 9080 wherein the agent is a kinesin antagonist.
9140. The method of item 9080 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9141. The method of item 9080 wherein the agent is an MAP kinase inhibitor.
9142. The method of item 9080 wherein the agent is a matrix metalloproteinase inhibitor.
9143. The method of item 9080 wherein the agent is an MCP- CCR2 inhibitor.
9144. The method of item 9080 wherein the agent is an mTOR inhibitor.
9145. The method of item 9080 wherein the agent is an mTOR kinase inhibitor. 9146. The method of item 9080 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
9147. The method of item 9080 wherein the agent is an MIF inhibitor.
9148. The method of item 9080 wherein the agent is an MMP inhibitor.
9149. The method of item 9080 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
9150. The method of item 9080 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9151. The method of item 9080 wherein the agent is a nitric oxide agonist.
9152. The method of item 9080 wherein the agent is an ornithine decarboxylase inhibitor.
9153. The method of item 9080 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
9154. The method of item 9080 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
9155. The method of item 9080 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN- 107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 9156. The method of item 9080 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9157. The method of item 9080 wherein the agent is a phosphatase inhibitor.
9158. The method of item 9080 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9159. The method of item 9080 wherein the agent is a PKC inhibitor.
9160. The method of item 9080 wherein the agent is a platelet activating factor antagonist.
9161. The method of item 9080 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
9162. The method of item 9080 wherein the agent is a prolyl hydroxylase inhibitor.
9163. The method of item 9080 wherein the agent is a polymorphonuclear neutrophil inhibitor.
9164. The method of item 9080 wherein the agent is a protein kinase B inhibitor.
9165. The method of item 9080 wherein the agent is a protein kinase C stimulant. 9166. The method of item 9080 wherein the agent is a purine nucleoside analogue.
9167. The method of item 9080 wherein the agent is a purinoreceptor P2X antagonist.
9168. The method of item 9080 wherein the agent is a Raf kinase inhibitor.
9169. The method of item 9080 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
9170. The method of item 9080 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
9171. The method of item 9080 wherein the agent is an SDF-1 antagonist.
9172. The method of item 9080 wherein the agent is a sheddase inhibitor.
9173. The method of item 9080 wherein the agent is an SRC inhibitor.
9174. The method of item 9080 wherein the agent is a stromelysin inhibitor.
9175. The method of item 9080 wherein the agent is an Syk kinase inhibitor.
9176. The method of item 9080 wherein the agent is a telomerase inhibitor.
9177. The method of item 9080 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9178. The method of item 9080 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9179. The method of item 9080 wherein the agent is a Toll receptor inhibitor.
9180. The method of item 9080 wherein the agent is a tubulin antagonist.
9181. The method of item 9080 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9182. The method of item 9080 wherein the agent is a VEGF inhibitor. 9183. The method of item 9080 wherein the agent is a vitamin D receptor agonist.
9184. The method of item 9080 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
9185. The method of item 9080 wherein the agent is AP-23573
(an mTOR inhibitor).
9186. The method of item 9080 wherein the agent is synthadotin (a tubulin antagonist).
9187. The method of item 9080 wherein the agent is S-0885 (a collagenase inhibitor).
9188. The method of item 9080 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
9189. The method of item 9080 wherein the agent is ixabepilone (an epithilone).
9190. The method of item 9080 wherein the agent is I DN-5390
(an angiogenesis inhibitor).
9191. The method of item 9080 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
9192. The method of item 9080 wherein the agent is ABT-518 (an angiogenesis inhibitor).
9193. The method of item 9080 wherein the agent is combretastatin (an angiogenesis inhibitor).
9194. The method of item 9080 wherein the agent is anecortave acetate (an angiogenesis inhibitor). 9195. The method of item 9080 wherein the agent is SB- 715992 (a kinesin antagonist).
9196. The method of item 9080 wherein the agent is temsirolimus (an mTOR inhibitor).
9197. The method of item 9080 wherein the agent is adalimumab (a TNFα antagonist).
9198. The method of item 9080, wherein the composition comprises a polymer.
9199. The method of item 9080, wherein the composition comprises a polymeric carrier.
9200. The method of item 9080 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
9201. The method of item 9080 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
9202. The method of item 9080 wherein the device has a coating that comprises the anti-scarring agent.
9203. The method of item 9080, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
9204. The method of item 9080, wherein the device has a coating that comprises the agent and directly contacts the sensor.
9205. The method of item 9080, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
9206. The method of item 9080, wherein the device has a coating that comprises the agent and partially covers the sensor. 9207. The method of item 9080, wherein the device has a coating that comprises the agent and completely covers the sensor.
9208. The method of item 9080, wherein the device has a uniform coating.
9209. The method of item 9080, wherein the device has a nonuniform coating.
9210. The method of item 9080, wherein the device has a discontinuous coating.
9211. The method of item 9080, wherein the device has a patterned coating.
9212. The method of item 9080, wherein the device has a coating with a thickness of 100 μm or less.
9213. The method of item 9080, wherein the device has a coating with a thickness of 10 μm or less.
9214. The method of item 9080, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
9215. The method of item 9080, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
9216. The method of item 9080, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
9217. The method of item 9080, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight. 9218. The method of item 9080, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
9219. The method of item 9080, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
9220. The method of item 9080, wherein the device has a coating, and wherein the coating further comprises a polymer.
9221. The method of item 9080, wherein the device has a first coating having a first composition and a second coating having a second composition.
9222. The method of item 9080, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
9223. The method of item 9080, wherein the composition comprises a polymer.
9224. The method of item 9080, wherein the composition comprises a polymeric carrier.
9225. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
9226. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. 9227. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
9228. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
9229. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
9230. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
9231. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
9232. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
9233. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
9234. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
9235. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer. 9236. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
9237. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
9238. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
9239. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
9240. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
9241. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
9242. The method of item 9080, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
9243. The method of item 9080 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
9244. The method of item 9080, wherein the device comprises a lubricious coating. 9245. The method of item 9080 wherein the anti-scarring agent is located within pores or holes of the device.
9246. The method of item 9080 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
9247. The method of item 9080, wherein the device comprises a second pharmaceutically active agent.
9248. The method of item 9080 wherein the device comprises an anti-inflammatory agent.
9249. The method of item 9080 wherein the device comprises an agent that inhibits infection.
9250. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
9251. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
9252. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
9253. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
9254. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
9255. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
9256. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate. 9257. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
9258. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
9259. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
9260. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
9261. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
9262. The method of item 9080 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
9263. The method of item 9080, further comprising an antithrombotic agent.
9264. The method of item 9080 wherein the device comprises a visualization agent.
9265. The method of item 9080 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
9266. The method of item 9080 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 9267. The method of item 9080 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
9268. The method of item 9080 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
9269. The method of item 9080 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
9270. The method of item 9080 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
9271. The method of item 9080 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
9272. The method of item 9080 wherein the device comprises an echogenic material.
9273. The method of item 9080 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
9274. The method of item 9080 wherein the device is sterile.
9275. The method of item 9080 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. 9276. The method of item 9080 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
9277. The method of item 9080 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
9278. The method of item 9080 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
9279. The method of item 9080 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
9280. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
9281. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
9282. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
9283. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
9284. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 9285. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
9286. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
9287. The method of item 9080 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
9288. The method of item 9080 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
9289. The method of item 9080 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
9290. The method of item 9080 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
9291. The method of item 9080 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
9292. The method of item 9080 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
9293. The method of item 9080 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9294. The method of item 9080 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 9295. The method of item 9080 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9296. The method of item 9080 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9297. The method of item 9080 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9298. The method of item 9080 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9299. The method of item 9080 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
9300. The method of item 9080 wherein the combining is performed by spraying the agent or the component onto the sensor.
9301. The method of item 9080 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
9302. The method of item 9080 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
9303. The method of item 9080 wherein the combining is performed by covalently attaching the agent or the composition to the sensor. 9304. The method of item 9080 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
9305. The method of item 9080 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
9306. The method of item 9080 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
9307. The method of item 9080 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
9308. The method of item 9080 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
9309. The method of item 9080 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
9310. The method of item 9080 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
9311. The method of item 9080 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
9312. The method of item 9080 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition. 9313. The method of item 9080 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
9314. The method of item 9080 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
9315. The method of item 9080 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
9316. The method of item 9080 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
9317. The method of item 9080 wherein the combining is performed by impregnating the sensor with the agent or the composition.
9318. The method of item 9080 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
9319. The method of item 9080 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
9320. The method of item 9080 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
9321. The method of item 9080 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor. 9322. The method of item 9080 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
9323. The method of item 9080 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
9324. The method of item 9080 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
9325. The method of item 9080 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
9326. The method of item 9080 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
9327. The method of item 9080 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
9328. The method of item 9080 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
9329. The method of item 9080 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
9330. The method of item 9080 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 9331. The method of any one of items 9080-9330 wherein the device is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site.
9332. A method for making a device comprising: combining a drug delivery implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
9333. The method of item 9332 wherein the agent is an adensosine A2A receptor antagonist.
9334. The method of item 9332 wherein the agent is an AKT inhibitor.
9335. The method of item 9332 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
9336. The method of item 9332 wherein the agent is an alpha 4 integrin antagonist.
9337. The method of item 9332 wherein the agent is an alpha 7 nicotinic receptor agonist.
9338. The method of item 9332 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE- 26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM- 1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2
Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (Med iQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA)1 NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
9339. The method of item 9332 wherein the agent is an apoptosis antagonist.
9340. The method of item 9332 wherein the agent is an apoptosis activator.
9341. The method of item 9332 wherein the agent is a beta 1 integrin antagonist.
9342. The method of item 9332 wherein the agent is a beta tubulin inhibitor. 9343. The method of item 9332 wherein the agent is a blocker of enzyme production in Hepatitis C.
9344. The method of item 9332 wherein the agent is a Bruton's tyrosine kinase inhibitor.
9345. The method of item 9332 wherein the agent is a calcineurin inhibitor.
9346. The method of item 9332 wherein the agent is a caspase 3 inhibitor.
9347. The method of item 9332 wherein the agent is a CC chemokine receptor antagonist.
9348. The method of item 9332 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
9349. The method of item 9332 wherein the agent is a cathepsin B inhibitor.
9350. The method of item 9332 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
9351. The method of item 9332 wherein the agent is a cathepsin L inhibitor.
9352. The method of item 9332 wherein the agent is a CD40 antagonist.
9353. The method of item 9332 wherein the agent is a chemokine receptor agonist. 9354. The method of item 9332 wherein the agent is a chymase inhibitor.
9355. The method of item 9332 wherein the agent is a collagenase antagonist.
9356. The method of item 9332 wherein the agent is a CXCR antagonist.
9357. The method of item 9332 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
9358. The method of item 9332 wherein the agent is a cyclooxygenase 1 inhibitor.
9359. The method of item 9332 wherein the agent is a DHFR inhibitor.
9360. The method of item 9332 wherein the agent is a dual integrin inhibitor.
9361. The method of item 9332 wherein the agent is an elastase inhibitor.
9362. The method of item 9332 wherein the agent is an elongation factor-1 alpha inhibitor. 9363. The method of item 9332 wherein the agent is an endothelial growth factor antagonist.
9364. The method of item 9332 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
9365. The method of item 9332 wherein the agent is an endotoxin antagonist.
9366. The method of item 9332 wherein the agent is an epothilone and tubulin binder.
9367. The method of item 9332 wherein the agent is an estrogen receptor antagonist.
9368. The method of item 9332 wherein the agent is an FGF inhibitor.
9369. The method of item 9332 wherein the agent is a farnexyl transferase inhibitor.
9370. The method of item 9332 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R)1 LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9371. The method of item 9332 wherein the agent is an FLT-3 kinase inhibitor.
9372. The method of item 9332 wherein the agent is an FGF receptor kinase inhibitor.
9373. The method of item 9332 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11 -9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9374. The method of item 9332 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV1 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
9375. The method of item 9332 wherein the agent is a histone deacetylase inhibitor.
9376. The method of item 9332 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 9377. The method of item 9332 wherein the agent is an ICAM inhibitor.
9378. The method of item 9332 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9379. The method of item 9332 wherein the agent is an IL-2 inhibitor.
9380. The method of item 9332 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9381. The method of item 9332 wherein the agent is an IMPDH (inosine monophosphate). 9382. The method of item 9332 wherein the agent is an integrin antagonist.
9383. The method of item 9332 wherein the agent is an interleukin antagonist.
9384. The method of item 9332 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
9385. The method of item 9332 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9386. The method of item 9332 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
9387. The method of item 9332 wherein the agent a JAK3 enzyme inhibitor.
9388. The method of item 9332 wherein the agent is a JNK inhibitor.
9389. The method of item 9332 wherein the agent is a kinase inhibitor.
9390. The method of item 9332 wherein the agent is kinesin antagonist.
9391. The method of item 9332 wherein the agent is a kinesin antagonist.
9392. The method of item 9332 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9393. The method of item 9332 wherein the agent is an MAP kinase inhibitor.
9394. The method of item 9332 wherein the agent is a matrix metalloproteinase inhibitor.
9395. The method of item 9332 wherein the agent is an MCP-
CCR2 inhibitor.
9396. The method of item 9332 wherein the agent is an mTOR inhibitor.
9397. The method of item 9332 wherein the agent is an mTOR kinase inhibitor.
9398. The method of item 9332 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
9399. The method of item 9332 wherein the agent is an MIF inhibitor.
9400. The method of item 9332 wherein the agent is an MMP inhibitor.
9401. The method of item 9332 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
9402. The method of item 9332 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 9403. The method of item 9332 wherein the agent is a nitric oxide agonist.
9404. The method of item 9332 wherein the agent is an ornithine decarboxylase inhibitor.
9405. The method of item 9332 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
9406. The method of item 9332 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
9407. The method of item 9332 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-
107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP- 860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
9408. The method of item 9332 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO- 5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9409. The method of item 9332 wherein the agent is a phosphatase inhibitor.
9410. The method of item 9332 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9411. The method of item 9332 wherein the agent is a PKC inhibitor.
9412. The method of item 9332 wherein the agent is a platelet activating factor antagonist.
9413. The method of item 9332 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
9414. The method of item 9332 wherein the agent is a prolyl hydroxylase inhibitor.
9415. The method of item 9332 wherein the agent is a polymorphonuclear neutrophil inhibitor.
9416. The method of item 9332 wherein the agent is a protein kinase B inhibitor.
9417. The method of item 9332 wherein the agent is a protein kinase C stimulant.
9418. The method of item 9332 wherein the agent is a purine nucleoside analogue.
9419. The method of item 9332 wherein the agent is a purinoreceptor P2X antagonist.
9420. The method of item 9332 wherein the agent is a Raf kinase inhibitor. 9421. The method of item 9332 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
9422. The method of item 9332 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
9423. The method of item 9332 wherein the agent is an SDF-1 antagonist.
9424. The method of item 9332 wherein the agent is a sheddase inhibitor.
9425. The method of item 9332 wherein the agent is an SRC inhibitor.
9426. The method of item 9332 wherein the agent is a stromelysin inhibitor.
9427. The method of item 9332 wherein the agent is an Syk kinase inhibitor.
9428. The method of item 9332 wherein the agent is a telomerase inhibitor.
9429. The method of item 9332 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9430. The method of item 9332 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab
(CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPFM 32294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9431. The method of item 9332 wherein the agent is a Toll receptor inhibitor.
9432. The method of item 9332 wherein the agent is a tubulin antagonist.
9433. The method of item 9332 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9434. The method of item 9332 wherein the agent is a VEGF inhibitor.
9435. The method of item 9332 wherein the agent is a vitamin D receptor agonist.
9436. The method of item 9332 wherein the agent is ZD-6474
(an angiogenesis inhibitor).
9437. The method of item 9332 wherein the agent is AP-23573 (an mTOR inhibitor). 9438. The method of item 9332 wherein the agent is synthadotin (a tubulin antagonist).
9439. The method of item 9332 wherein the agent is S-0885 (a collagenase inhibitor).
9440. The method of item 9332 wherein the agent is aplidine
(an elongation factor-1 alpha inhibitor).
9441. The method of item 9332 wherein the agent is ixabepilone (an epithilone).
9442. The method of item 9332 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
9443. The method of item 9332 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
9444. The method of item 9332 wherein the agent is ABT-518 (an angiogenesis inhibitor).
9445. The method of item 9332 wherein the agent is combretastatin (an angiogenesis inhibitor).
9446. The method of item 9332 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
9447. The method of item 9332 wherein the agent is SB- 715992 (a kinesin antagonist).
9448. The method of item 9332 wherein the agent is temsirolimus (an mTOR inhibitor).
9449. The method of item 9332 wherein the agent is adalimumab (a TNFα antagonist). 9450. The method of item 9332, wherein the composition comprises a polymer.
9451. The method of item 9332, wherein the composition comprises a polymeric carrier.
9452. The method of item 9332 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
9453. The method of item 9332 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
9454. The method of item 9332 wherein the device has a coating that comprises the anti-scarring agent.
9455. The method of item 9332, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
9456. The method of item 9332, wherein the device has a coating that comprises the agent and directly contacts the sensor.
9457. The method of item 9332, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
9458. The method of item 9332, wherein the device has a coating that comprises the agent and partially covers the sensor.
9459. The method of item 9332, wherein the device has a coating that comprises the agent and completely covers the sensor.
9460. The method of item 9332, wherein the device has a uniform coating.
9461. The method of item 9332, wherein the device has a non- uniform coating. 9462. The method of item 9332, wherein the device has a discontinuous coating.
9463. The method of item 9332, wherein the device has a patterned coating.
9464. The method of item 9332, wherein the device has a coating with a thickness of 100 μm or less.
9465. The method of item 9332, wherein the device has a coating with a thickness of 10 μm or less.
9466. The method of item 9332, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
9467. The method of item 9332, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
9468. The method of item 9332, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
9469. The method of item 9332, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
9470. The method of item 9332, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
9471. The method of item 9332, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 9472. The method of item 9332, wherein the device has a coating, and wherein the coating further comprises a polymer.
9473. The method of item 9332, wherein the device has a first coating having a first composition and a second coating having a second composition.
9474. The method of item 9332, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
9475. The method of item 9332, wherein the composition comprises a polymer.
9476. The method of item 9332, wherein the composition comprises a polymeric carrier.
9477. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
9478. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
9479. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
9480. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 9481. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
9482. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
9483. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
9484. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
9485. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
9486. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
9487. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
9488. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
9489. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 9490. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
9491. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
9492. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
9493. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
9494. The method of item 9332, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
9495. The method of item 9332 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
9496. The method of item 9332, wherein the device comprises a lubricious coating.
9497. The method of item 9332 wherein the anti-scarring agent is located within pores or holes of the device.
9498. The method of item 9332 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
9499. The method of item 9332, wherein the device comprises a second pharmaceutically active agent. 9500. The method of item 9332 wherein the device comprises an anti-inflammatory agent.
9501. The method of item 9332 wherein the device comprises an agent that inhibits infection.
9502. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
9503. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
9504. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
9505. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
9506. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
9507. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
9508. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
9509. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
9510. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
9511. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 9512. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
9513. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
9514. The method of item 9332 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
9515. The method of item 9332, further comprising an antithrombotic agent.
9516. The method of item 9332 wherein the device comprises a visualization agent.
9517. The method of item 9332 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
9518. The method of item 9332 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
9519. The method of item 9332 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
9520. The method of item 9332 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 9521. The method of item 9332 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
9522. The method of item 9332 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
9523. The method of item 9332 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
9524. The method of item 9332 wherein the device comprises an echogenic material.
9525. The method of item 9332 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
9526. The method of item 9332 wherein the device is sterile.
9527. The method of item 9332 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
9528. The method of item 9332 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
9529. The method of item 9332 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 9530. The method of item 9332 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
9531. The method of item 9332 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
9532. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
9533. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
9534. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
9535. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
9536. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
9537. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
9538. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 9539. The method of item 9332 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
9540. The method of item 9332 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
9541. The method of item 9332 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
9542. The method of item 9332 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
9543. The method of item 9332 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
9544. The method of item 9332 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
9545. The method of item 9332 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9546. The method of item 9332 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9547. The method of item 9332 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9548. The method of item 9332 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 9549. The method of item 9332 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9550. The method of item 9332 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
9551. The method of item 9332 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
9552. The method of item 9332 wherein the combining is performed by spraying the agent or the component onto the sensor.
9553. The method of item 9332 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
9554. The method of item 9332 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
9555. The method of item 9332 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
9556. The method of item 9332 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor.
9557. The method of item 9332 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
9558. The method of item 9332 wherein the combining is performed by coating the sensor with a substance that absorbs the agent. 9559. The method of item 9332 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
9560. The method of item 9332 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
9561. The method of item 9332 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
9562. The method of item 9332 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
9563. The method of item 9332 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
9564. The method of item 9332 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition.
9565. The method of item 9332 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
9566. The method of item 9332 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
9567. The method of item 9332 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition. 9568. The method of item 9332 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
9569. The method of item 9332 wherein the combining is performed by impregnating the sensor with the agent or the composition.
9570. The method of item 9332 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
9571. The method of item 9332 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
9572. The method of item 9332 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
9573. The method of item 9332 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
9574. The method of item 9332 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
9575. The method of item 9332 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
9576. The method of item 9332 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor. 9577. The method of item 9332 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
9578. The method of item 9332 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
9579. The method of item 9332 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
9580. The method of item 9332 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
9581. The method of item 9332 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
9582. The method of item 9332 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
9583. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is sensor. 9584. The method for implanting a medical device according to item 9583 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
9585. The method for implanting a medical device according to item 9583 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
9586. The method for implanting a medical device according to item 9583 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
9587. The method for implanting a medical device according to item 9583 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
9588. The method for implanting a medical device according to item 9583 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
9589. The method for implanting a medical device according to item 9583 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
9590. The method of item 9583 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist. 9591. The method of item 9583 wherein the anti-fibrotic agent is an AKT inhibitor.
9592. The method of item 9583 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
9593. The method of item 9583 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
9594. The method of item 9583 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
9595. The method of item 9583 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BiBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW- 114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
9596. The method of item 9583 wherein the anti-fibrotic agent is an apoptosis antagonist.
9597. The method of item 9583 wherein the anti-fibrotic agent is an apoptosis activator.
9598. The method of item 9583 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
9599. The method of item 9583 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
9600. The method of item 9583 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
9601. The method of item 9583 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
9602. The method of item 9583 wherein the anti-fibrotic agent is a calcineurin inhibitor.
9603. The method of item 9583 wherein the anti-fibrotic agent is a caspase 3 inhibitor. 9604. The method of item 9583 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
9605. The method of item 9583 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
9606. The method of item 9583 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
9607. The method of item 9583 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
9608. The method of item 9583 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
9609. The method of item 9583 wherein the anti-fibrotic agent is a CD40 antagonist.
9610. The method of item 9583 wherein the anti-fibrotic agent is a chemokine receptor agonist.
9611. The method of item 9583 wherein the anti-fibrotic agent is a chymase inhibitor.
9612. The method of item 9583 wherein the anti-fibrotic agent is a collagenase antagonist.
9613. The method of item 9583 wherein the anti-fibrotic agent is a CXCR antagonist.
9614. The method of item 9583 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a
CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
9615. The method of item 9583 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
9616. The method of item 9583 wherein the anti-fibrotic agent is a DHFR inhibitor.
9617. The method of item 9583 wherein the anti-fibrotic agent is a dual integrin inhibitor.
9618. The method of item 9583 wherein the anti-fibrotic agent is an elastase inhibitor.
9619. The method of item 9583 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
9620. The method of item 9583 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
9621. The method of item 9583 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
9622. The method of item 9583 wherein the anti-fibrotic agent is an endotoxin antagonist.
9623. The method of item 9583 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
9624. The method of item 9583 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
9625. The method of item 9583 wherein the anti-fibrotic agent is an FGF inhibitor.
9626. The method of item 9583 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor.
9627. The method of item 9583 wherein the anti-fibrotic agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9628. The method of item 9583 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
9629. The method of item 9583 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
9630. The method of item 9583 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9631. The method of item 9583 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
9632. The method of item 9583 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
9633. The method of item 9583 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9634. The method of item 9583 wherein the anti-fibrotic agent is an ICAM inhibitor.
9635. The method of item 9583 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9636. The method of item 9583 wherein the anti-fibrotic agent is an IL-2 inhibitor.
9637. The method of item 9583 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9638. The method of item 9583 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
9639. The method of item 9583 wherein the anti-fibrotic agent is an integrin antagonist.
9640. The method of item 9583 wherein the anti-fibrotic agent is an interleukin antagonist.
9641. The method of item 9583 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
9642. The method of item 9583 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9643. The method of item 9583 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor. 9644. The method of item 9583 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
9645. The method of item 9583 wherein the anti-fibrotic agent is a JNK inhibitor.
9646. The method of item 9583 wherein the anti-fibrotic agent is a kinase inhibitor.
9647. The method of item 9583 wherein the anti-fibrotic agent is kinesin antagonist.
9648. The method of item 9583 wherein the anti-fibrotic agent is a kinesin antagonist.
9649. The method of item 9583 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), Ieucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9650. The method of item 9583 wherein the anti-fibrotic agent is an MAP kinase inhibitor. 9651. The method of item 9583 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
9652. The method of item 9583 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
9653. The method of item 9583 wherein the anti-fibrotic agent is an mTOR inhibitor.
9654. The method of item 9583 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
9655. The method of item 9583 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
9656. The method of item 9583 wherein the anti-fibrotic agent is an MIF inhibitor.
9657. The method of item 9583 wherein the anti-fibrotic agent is an MMP inhibitor.
9658. The method of item 9583 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
9659. The method of item 9583 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9660. The method of item 9583 wherein the anti-fibrotic agent is a nitric oxide agonist.
9661. The method of item 9583 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
9662. The method of item 9583 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof. 9663. The method of item 9583 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
9664. The method of item 9583 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
9665. The method of item 9583 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4),
ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from
CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9666. The method of item 9583 wherein the anti-fibrotic agent is a phosphatase inhibitor.
9667. The method of item 9583 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC:3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPFM 17658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9668. The method of item 9583 wherein the anti-fibrotic agent is a PKC inhibitor.
9669. The method of item 9583 wherein the anti-fibrotic agent is a platelet activating factor antagonist. 9670. The method of item 9583 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
9671. The method of item 9583 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
9672. The method of item 9583 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
9673. The method of item 9583 wherein the anti-fibrotic agent is a protein kinase B inhibitor.
9674. The method of item 9583 wherein the anti-fibrotic agent is a protein kinase C stimulant.
9675. The method of item 9583 wherein the anti-fibrotic agent is a purine nucleoside analogue.
9676. The method of item 9583 wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist.
9677. The method of item 9583 wherein the anti-fibrotic agent is a Raf kinase inhibitor.
9678. The method of item 9583 wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
9679. The method of item 9583 wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
9680. The method of item 9583 wherein the anti-fibrotic agent is an SDF-1 antagonist.
9681. The method of item 9583 wherein the anti-fibrotic agent is a sheddase inhibitor. 9682. The method of item 9583 wherein the anti-fibrotic agent is an SRC inhibitor.
9683. The method of item 9583 wherein the anti-fibrotic agent is a stromelysin inhibitor.
9684. The method of item 9583 wherein the anti-fibrotic agent is an Syk kinase inhibitor.
9685. The method of item 9583 wherein the anti-fibrotic agent is a telomerase inhibitor.
9686. The method of item 9583 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No.
53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9687. The method of item 9583 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalϊmumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9688. The method of item 9583 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
9689. The method of item 9583 wherein the anti-fibrotic agent is a tubulin antagonist. 9690. The method of item 9583 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9691. The method of item 9583 wherein the anti-fibrotic agent is a VEGF inhibitor.
9692. The method of item 9583 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
9693. The method of item 9583 wherein the anti-fibrotic agent is
ZD-6474 (an angiogenesis inhibitor).
9694. The method of item 9583 wherein the anti-fibrotic agent is AP-23573 (an mTOR inhibitor).
9695. The method of item 9583 wherein the anti-fibrotic agent is synthadotin (a tubulin antagonist).
9696. The method of item 9583 wherein the anti-fibrotic agent is S-0885 (a collagenase inhibitor).
9697. The method of item 9583 wherein the anti-fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
9698. The method of item 9583 wherein the anti-fibrotic agent is ixabepilone (an epithilone). 9699. The method of item 9583 wherein the anti-fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
9700. The method of item 9583 wherein the anti-fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
9701. The method of item 9583 wherein the anti-fibrotic agent is
ABT-518 (an angiogenesis inhibitor).
9702. The method of item 9583 wherein the anti-fibrotic agent is combretastatin (an angiogenesis inhibitor).
9703. The method of item 9583 wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
9704. The method of item 9583 wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
9705. The method of item 9583 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
9706. The method of item 9583 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
9707. The method of item 9583 wherein the anti-infective agent is an anthracycline.
9708. The method of item 9583 wherein the anti-infective agent is doxorubicin.
9709. The method of item 9583 wherein the anti-infective agent is mitoxantrone.
9710. The method of item 9583 wherein the anti-infective agent is a fluoropyrimidine. 9711. The method of item 9583 wherein the anti-infective agent is 5-fluorouracil (5-FU).
9712. The method of item 9583 wherein the anti-infective agent is a folic acid antagonist.
9713. The method of item 9583 wherein the anti-infective agent is methotrexate.
9714. The method of item 9583 wherein the anti-infective agent is a podophylotoxin.
9715. The method of item 9583 wherein the anti-infective agent is etoposide.
9716. The method of item 9583 wherein the anti-infective agent is camptothecin.
9717. The method of item 9583 wherein the anti-infective agent is a hydroxyurea.
9718. The method of item 9583 wherein the anti-infective agent is a platinum complex.
9719. The method of item 9583 wherein the anti-infective agent is cisplatin.
9720. The method of item 9583 wherein the composition comprises an anti-thrombotic agent.
9721. The method of item 9583 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
9722. The method of item 9583 wherein the polymer is formed from reactants comprising protein. 006/011726
9723. The method of item 9583 wherein the polymer is formed from reactants comprising carbohydrate.
9724. The method of item 9583 wherein the polymer is formed from reactants comprising biodegradable polymer.
9725. The method of item 9583 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
9726. The method of item 9583 wherein the polymer is formed from reactants comprising collagen.
9727. The method of item 9583 wherein the polymer is formed from reactants comprising methylated collagen.
9728. The method of item 9583 wherein the polymer is formed from reactants comprising fibrinogen.
9729. The method of item 9583 wherein the polymer is formed from reactants comprising thrombin.
9730. The method of item 9583 wherein the polymer is formed from reactants comprising blood plasma.
9731. The method of item 9583 wherein the polymer is formed from reactants comprising calcium salt.
9732. The method of item 9583 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
9733. The method of item 9583 wherein the polymer is formed from reactants comprising fibrinogen analog.
9734. The method of item 9583 wherein the polymer is formed from reactants comprising albumin. 9735. The method of item 9583 wherein the polymer is formed from reactants comprising plasminogen.
9736. The method of item 9583 wherein the polymer is formed from reactants comprising von Willebrands factor.
9737. The method of item 9583 wherein the polymer is formed from reactants comprising Factor VIII.
9738. The method of item 9583 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
9739. The method of item 9583 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
9740. The method of item 9583 wherein the polymer is formed from reactants comprising telopeptide collagen.
9741. The method of item 9583 wherein the polymer is formed from reactants comprising crosslinked collagen.
9742. The method of item 9583 wherein the polymer is formed from reactants comprising aprotinin.
9743. The method of item 9583 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
9744. The method of item 9583 wherein the polymer is formed from reactants comprising gelatin.
9745. The method of item 9583 wherein the polymer is formed from reactants comprising protein conjugates.
9746. The method of item 9583 wherein the polymer is formed from reactants comprising gelatin conjugates. 9747. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic polymer.
9748. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
9749. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
9750. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
9751. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
9752. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
9753. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
9754. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
9755. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
9756. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 9757. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
9758. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
9759. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
9760. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
9761. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
9762. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
9763. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
9764. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
9765. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups. 9766. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
9767. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
9768. The method of item 9583 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
9769. The method of item 9583 wherein the polymer is formed from reactants comprising polylysine.
9770. The method of item 9583 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
9771. The method of item 9583 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
9772. The method of item 9583 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
9773. The method of item 9583 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
9774. The method of item 9583 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 9775. The method of item 9583 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
9776. The method of item 9583 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
9777. The method of item 9583 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
9778. The method of item 9583 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
9779. The method of item 9583 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
9780. The method of item 9583 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
9781. The method of item 9583 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
9782. The method of item 9583 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
9783. The method of item 9583 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine. 9784. The method of item 9583 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
9785. The method of item 9583 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
9786. The method of item 9583 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
9787. The method of item 9583 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
9788. The method of item 9583 wherein the polymer is formed from reactants comprising hyaluronic acid.
9789. The method of item 9583 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
9790. The method of item 9583 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
9791. The method of item 9583 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
9792. The method of item 9583 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
9793. The method of item 9583 wherein the composition comprises a colorant.
9794. The method of item 9583 wherein the composition is sterile.
9795. The method of any one of items 9583-9794 wherein the sensor is a blood or tissue glucose monitor.
9796. The method of any one of items 9583-9794 wherein the sensor is an electrolyte sensor.
9797. The method of any one of items 9583-9794 wherein the sensor is a blood constituent sensor.
9798. The method of any one of items 9583-9794 wherein the sensor is a temperature sensor.
9799. The method of any one of items 9583-9794 wherein the sensor is a pH sensor.
9800. The method of any one of items 9583-9794 wherein the sensor is an optical sensor.
9801. The method of any one of items 9583-9794 wherein the sensor is an amperometric sensor.
9802. The method of any one of items 9583-9794 wherein the sensor is a pressure sensor.
9803. The method of any one of items 9583-9794 wherein the sensor is a biosensor. 9804. The method of any one of items 9583-9794 wherein the sensor is a sensing transponder.
9805. The method of any one of items 9583-9794 wherein the sensor is a strain sensor.
9806. The method of any one of items 9583-9794 wherein the sensor is a magnetoresistive sensor.
9807. The method of any one of items 9583-9794 wherein the sensor is a cardiac sensor.
9808. The method of any one of items 9583-9794 wherein the sensor is a respiratory sensor.
9809. The method of any one of items 9583-9794 wherein the sensor is an auditory sensor.
9810. The method of any one of items 9583-9794 wherein the sensor is a metabolite sensor.
9811. The method of any one of items 9583-9794 wherein the sensor detects mechanical changes.
9812. The method of any one of items 9583-9794 wherein the sensor detects physical changes.
9813. The method of any one of items 9583-9794 wherein the sensor detects electrochemical changes.
9814. The method of any one of items 9583-9794 wherein the sensor detects magnetic changes.
9815. The method of any one of items 9583-9794 wherein the sensor detects acceleration changes. 9816. The method of any one of items 9583-9794 wherein the sensor detects ionizing radiation changes.
9817. The method of any one of items 9583-9794 wherein the sensor detects acoustic wave changes.
9818. The method of any one of items 9583-9794 wherein the sensor detects chemical changes.
9819. The method of any one of items 9583-9794 wherein the sensor detects drug concentration changes.
9820. The method of any one of items 9583-9794 wherein the sensor detects hormone changes.
9821. The method of any one of items 9583-9794 wherein the sensor detects barometric changes.
9822. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a blood or tissue glucose monitor.
9823. The method for implanting a medical device according to item 9822 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
9824. The method for implanting a medical device according to item 9822 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
9825. The method for implanting a medical device according to item 9822 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
9826. The method for implanting a medical device according to item 9822 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
9827. The method for implanting a medical device according to item 9822 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
9828. The method for implanting a medical device according to item 9822 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
9829. The method of item 9822 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
9830. The method of item 9822 wherein the anti-fibrotic agent is an AKT inhibitor.
9831. The method of item 9822 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is
Pharmaprojects No. 5754 (Merck KgaA). 9832. The method of item 9822 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
9833. The method of item 9822 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
9834. The method of item 9822 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
9835. The method of item 9822 wherein the anti-fibrotic agent is an apoptosis antagonist.
9836. The method of item 9822 wherein the anti-fibrotic agent is an apoptosis activator.
9837. The method of item 9822 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
9838. The method of item 9822 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
9839. The method of item 9822 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
9840. The method of item 9822 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
9841. The method of item 9822 wherein the anti-fibrotic agent is a calcineurin inhibitor.
9842. The method of item 9822 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
9843. The method of item 9822 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
9844. The method of item 9822 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 9845. The method of item 9822 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
9846. The method of item 9822 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
9847. The method of item 9822 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
9848. The method of item 9822 wherein the anti-fibrotic agent is a CD40 antagonist.
9849. The method of item 9822 wherein the anti-fibrotic agent is a chemokine receptor agonist.
9850. The method of item 9822 wherein the anti-fibrotic agent is a chymase inhibitor.
9851. The method of item 9822 wherein the anti-fibrotic agent is a collagenase antagonist.
9852. The method of item 9822 wherein the anti-fibrotic agent is a CXCR antagonist.
9853. The method of item 9822 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a
CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
9854. The method of item 9822 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
9855. The method of item 9822 wherein the anti-fibrotic agent is a DHFR inhibitor.
9856. The method of item 9822 wherein the anti-fibrotic agent is a dual integrin inhibitor.
9857. The method of item 9822 wherein the anti-fibrotic agent is an elastase inhibitor.
9858. The method of item 9822 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
9859. The method of item 9822 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
9860. The method of item 9822 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
9861. The method of item 9822 wherein the anti-fibrotic agent is an endotoxin antagonist. 9862. The method of item 9822 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
9863. The method of item 9822 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
9864. The method of item 9822 wherein the anti-fibrotic agent is an FGF inhibitor.
9865. The method of item 9822 wherein the anti-fibrotic agent is a famexyl transferase inhibitor.
9866. The method of item 9822 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9867. The method of item 9822 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
9868. The method of item 9822 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
9869. The method of item 9822 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201
(Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9870. The method of item 9822 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)- 1-OXO-), and an analogue or derivative thereof.
9871. The method of item 9822 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
9872. The method of item 9822 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9873. The method of item 9822 wherein the anti-fibrotic agent is an ICAM inhibitor.
9874. The method of item 9822 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-
424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9875. The method of item 9822 wherein the anti-fibrotic agent is an IL-2 inhibitor.
9876. The method of item 9822 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide
(Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemϊa, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9877. The method of item 9822 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
9878. The method of item 9822 wherein the anti-fibrotic agent is an integrin antagonist.
9879. The method of item 9822 wherein the anti-fibrotic agent is an interleukin antagonist.
9880. The method of item 9822 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
9881. The method of item 9822 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9882. The method of item 9822 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
9883. The method of item 9822 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
9884. The method of item 9822 wherein the anti-fibrotic agent is a JNK inhibitor.
9885. The method of item 9822 wherein the anti-fibrotic agent is a kinase inhibitor. 9886. The method of item 9822 wherein the anti-fibrotic agent is kinesin antagonist.
9887. The method of item 9822 wherein the anti-fibrotic agent is a kinesin antagonist.
9888. The method of item 9822 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9889. The method of item 9822 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
9890. The method of item 9822 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
9891. The method of item 9822 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
9892. The method of item 9822 wherein the anti-fibrotic agent is an mTOR inhibitor. 9893. The method of item 9822 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
9894. The method of item 9822 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
9895. The method of item 9822 wherein the anti-fibrotic agent is an MIF inhibitor.
9896. The method of item 9822 wherein the anti-fibrotic agent is an MMP inhibitor.
9897. The method of item 9822 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin
(CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR- 240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 9898. The method of item 9822 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9899. The method of item 9822 wherein the anti-fibrotic agent is a nitric oxide agonist.
9900. The method of item 9822 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
9901. The method of item 9822 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
9902. The method of item 9822 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
9903. The method of item 9822 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-
993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
9904. The method of item 9822 wherein the anti-fibrotic agent is
(-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735
(GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus
Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0)
(GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9905. The method of item 9822 wherein the anti-fibrotic agent is a phosphatase inhibitor.
9906. The method of item 9822 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1 ), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9907. The method of item 9822 wherein the anti-fibrotic agent is a PKC inhibitor.
9908. The method of item 9822 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
9909. The method of item 9822 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
9910. The method of item 9822 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
9911. The method of item 9822 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor. 9912. The method of item 9822 wherein the anti-fibrotic agent is a protein kinase B inhibitor. <
9913. The method of item 9822 wherein the anti-fibrotic agent is a protein kinase C stimulant.
I 9914. The method of item 9822 wherein the anti-fibrotic agent is a purine nucleoside analogue.
9915. The method of item 9822 wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist.
9916. The method of item 9822 wherein the anti-fibrotic agent is a Raf kinase inhibitor.
9917. The method of item 9822 wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
9918. The method of item 9822 wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
9919. The method of item 9822 wherein the anti-fibrotic agent is an SDF-1 antagonist.
9920. The method of item 9822 wherein the anti-fibrotic agent is a sheddase inhibitor.
9921. The method of item 9822 wherein the anti-fibrotic agent is an SRC inhibitor.
9922. The method of item 9822 wherein the anti-fibrotic agent is a stromelysin inhibitor.
9923. The method of item 9822 wherein the anti-fibrotic agent is an Syk kinase inhibitor. 9924. The method of item 9822 wherein the anti-fibrotic agent is a telomerase inhibitor.
9925. The method of item 9822 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9926. The method of item 9822 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9927. The method of item 9822 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
9928. The method of item 9822 wherein the anti-fibrotic agent is a tubulin antagonist.
9929. The method of item 9822 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9930. The method of item 9822 wherein the anti-fibrotic agent is a VEGF inhibitor.
9931. The method of item 9822 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
9932. The method of item 9822 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
9933. The method of item 9822 wherein the anti-fibrotic agent is AP-23573 (an mTOR inhibitor).
9934. The method of item 9822 wherein the anti-fibrotic agent is synthadotin (a tubulin antagonist).
9935. The method of item 9822 wherein the anti-fibrotic agent is S-0885 (a collagenase inhibitor).
9936. The method of item 9822 wherein the anti-fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
9937. The method of item 9822 wherein the anti-fibrotic agent is ixabepilone (an epithilone).
9938. The method of item 9822 wherein the anti-fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
9939. The method of item 9822 wherein the anti-fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
9940. The method of item 9822 wherein the anti-fibrotic agent is ABT-518 (an angiogenesis inhibitor). 9941. The method of item 9822 wherein the anti-fibrotic agent is combretastatin (an angiogenesis inhibitor).
9942. The method of item 9822 wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
9943. The method of item 9822 wherein the anti-fibrotic agent is
SB-715992 (a kinesin antagonist).
9944. The method of item 9822 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
9945. The method of item 9822 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
9946. The method of item 9822 wherein the anti-infective agent is an anthracycline.
9947. The method of item 9822 wherein the anti-infective agent is doxorubicin.
9948. The method of item 9822 wherein the anti-infective agent is mitoxantrone.
9949. The method of item 9822 wherein the anti-infective agent is a fluoropyrimidine.
9950. The method of item 9822 wherein the anti-infective agent is 5-fluorouracil (5-FU).
9951. The method of item 9822 wherein the anti-infective agent is a folic acid antagonist.
9952. The method of item 9822 wherein the anti-infective agent is methotrexate. 9953. The method of item 9822 wherein the anti-infective agent is a podophylotoxin.
9954. The method of item 9822 wherein the anti-infective agent is etoposide.
9955. The method of item 9822 wherein the anti-infective agent is camptothecin.
9956. The method of item 9822 wherein the anti-infective agent is a hydroxyurea.
9957. The method of item 9822 wherein the anti-infective agent is a platinum complex.
9958. The method of item 9822 wherein the anti-infective agent is cisplatin.
9959. The method of item 9822 wherein the composition comprises an anti-thrombotic agent.
9960. The method of item 9822 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
9961. The method of item 9822 wherein the polymer is formed from reactants comprising protein.
9962. The method of item 9822 wherein the polymer is formed from reactants comprising carbohydrate.
9963. The method of item 9822 wherein the polymer is formed from reactants comprising biodegradable polymer.
9964. The method of item 9822 wherein the polymer is formed from reactants comprising nonbiodegradable polymer. 9965. The method of item 9822 wherein the polymer is formed from reactants comprising collagen.
9966. The method of item 9822 wherein the polymer is formed from reactants comprising methylated collagen.
9967. The method of item 9822 wherein the polymer is formed from reactants comprising fibrinogen.
9968. The method of item 9822 wherein the polymer is formed from reactants comprising thrombin.
9969. The method of item 9822 wherein the polymer is formed from reactants comprising blood plasma.
9970. The method of item 9822 wherein the polymer is formed from reactants comprising calcium salt.
9971. The method of item 9822 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
9972. The method of item 9822 wherein the polymer is formed from reactants comprising fibrinogen analog.
9973. The method of item 9822 wherein the polymer is formed from reactants comprising albumin.
9974. The method of item 9822 wherein the polymer is formed from reactants comprising plasminogen.
9975. The method of item 9822 wherein the polymer is formed from reactants comprising von Willebrands factor.
9976. The method of item 9822 wherein the polymer is formed from reactants comprising Factor VIII. 9977. The method of item 9822 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
9978. The method of item 9822 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
9979. The method of item 9822 wherein the polymer is formed from reactants comprising telopeptide collagen.
9980. The method of item 9822 wherein the polymer is formed from reactants comprising crosslinked collagen.
9981. The method of item 9822 wherein the polymer is formed from reactants comprising aprotinin.
9982. The method of item 9822 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
9983. The method of item 9822 wherein the polymer is formed from reactants comprising gelatin.
9984. The method of item 9822 wherein the polymer is formed from reactants comprising protein conjugates.
9985. The method of item 9822 wherein the polymer is formed from reactants comprising gelatin conjugates.
9986. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic polymer.
9987. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
9988. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 9989. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
9990. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
9991. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
9992. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
9993. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
9994. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
9995. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
9996. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
9997. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 9998. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
9999. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
10000. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
10001. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
10002. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
10003. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
10004. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
10005. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
10006. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide. 10007. The method of item 9822 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
10008. The method of item 9822 wherein the polymer is formed from reactants comprising polylysine.
10009. The method of item 9822 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
10010. The method of item 9822 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
10011. The method of item 9822 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
10012. The method of item 9822 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
10013. The method of item 9822 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10014. The method of item 9822 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10015. The method of item 9822 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 10016. The method of item 9822 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
10017. The method of item 9822 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
10018. The method of item 9822 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
10019. The method of item 9822 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10020. The method of item 9822 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10021. The method of item 9822 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
10022. The method of item 9822 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
10023. The method of item 9822 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
10024. The method of item 9822 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups. 10025. The method of item 9822 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10026. The method of item 9822 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10027. The method of item 9822 wherein the polymer is formed from reactants comprising hyaluronic acid.
10028. The method of item 9822 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
10029. The method of item 9822 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
10030. The method of item 9822 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
10031. The method of item 9822 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
10032. The method of item 9822 wherein the composition comprises a colorant.
10033. The method of item 9822 wherein the composition is sterile. 10034. The method of any one of items 9822-10033 wherein the device is deliverable to the vascular system transluminally using a catheter on a stent platform.
10035. The method of any one of items 9822-10033 wherein the device is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
10036. The method of any one of items 9822-10033 wherein the device is an electrode composed of an analyte responsive enzyme.
10037. The method of any one of items 9822-10033 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
10038. The method of any one of items 9822-10033 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin.
10039. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is pressure or stress sensor.
10040. The method for implanting a medical device according to item 10039 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
10041. The method for implanting a medical device according to item 10039 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
10042. The method for implanting a medical device according to item 10039 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
10043. The method for implanting a medical device according to item 10039 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host.
10044. The method for implanting a medical device according to item 10039 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
10045. The method for implanting a medical device according to item 10039 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
10046. The method of item 10039 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist. 10047. The method of item 10039 wherein the anti-fibrotic agent is an AKT inhibitor.
10048. The method of item 10039 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10049. The method of item 10039 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
10050. The method of item 10039 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
10051. The method of item 10039 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TlE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004
(Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10052. The method of item 10039 wherein the anti-fibrotic agent is an apoptosis antagonist.
10053. The method of item 10039 wherein the anti-fibrotic agent is an apoptosis activator.
10054. The method of item 10039 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
10055. The method of item 10039 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
10056. The method of item 10039 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
10057. The method of item 10039 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
10058. The method of item 10039 wherein the anti-fibrotic agent is a calcineurin inhibitor.
10059. The method of item 10039 wherein the anti-fibrotic agent is a caspase 3 inhibitor. 10060. The method of item 10039 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
10061. The method of item 10039 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10062. The method of item 10039 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
10063. The method of item 10039 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10064. The method of item 10039 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
10065. The method of item 10039 wherein the anti-fibrotic agent is a CD40 antagonist.
10066. The method of item 10039 wherein the anti-fibrotic agent is a chemokine receptor agonist.
10067. The method of item 10039 wherein the anti-fibrotic agent is a chymase inhibitor.
10068. The method of item 10039 wherein the anti-fibrotic agent is a collagenase antagonist.
10069. The method of item 10039 wherein the anti-fibrotic agent is a CXCR antagonist.
10070. The method of item 10039 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a
CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
10071. The method of item 10039 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
10072. The method of item 10039 wherein the anti-fibrotic agent is a DHFR inhibitor.
10073. The method of item 10039 wherein the anti-fibrotic agent is a dual integrin inhibitor.
10074. The method of item 10039 wherein the anti-fibrotic agent is an elastase inhibitor.
10075. The method of item 10039 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
10076. The method of item 10039 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
10077. The method of item 10039 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652
(GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10078. The method of item 10039 wherein the anti-fibrotic agent is an endotoxin antagonist.
10079. The method of item 10039 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
10080. The method of item 10039 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
10081. The method of item 10039 wherein the anti-fibrotic agent is an FGF inhibitor.
10082. The method of item 10039 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor.
10083. The method of item 10039 wherein the anti-fibrotic agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10084. The method of item 10039 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
10085. The method of item 10039 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
10086. The method of item 10039 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10087. The method of item 10039 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyi)-1-oxo-), and an analogue or derivative thereof.
10088. The method of item 10039 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
10089. The method of item 10039 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10090. The method of item 10039 wherein the anti-fibrotic agent is an ICAM inhibitor.
10091. The method of item 10039 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10092. The method of item 10039 wherein the anti-fibrotic agent is an IL-2 inhibitor.
10093. The method of item 10039 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
10094. The method of item 10039 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
10095. The method of item 10039 wherein the anti-fibrotic agent is an integrin antagonist.
10096. The method of item 10039 wherein the anti-fibrotic agent is an interleukin antagonist.
10097. The method of item 10039 wherein the anti-fibrotic agent is an inhibitor of type Ml receptor tyrosine kinase.
10098. The method of item 10039 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
10099. The method of item 10039 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor. 10100. The method of item 10039 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
10101. The method of item 10039 wherein the anti-fibrotic agent is a JNK inhibitor.
10102. The method of item 10039 wherein the anti-fibrotic agent is a kinase inhibitor.
10103. The method of item 10039 wherein the anti-fibrotic agent is kinesin antagonist.
10104. The method of item 10039 wherein the anti-fibrotic agent is a kinesin antagonist.
10105. The method of item 10039 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10106. The method of item 10039 wherein the anti-fibrotic agent is an MAP kinase inhibitor. 10107. The method of item 10039 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
10108. The method of item 10039 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
10109. The method of item 10039 wherein the anti-fibrotic agent is an mTOR inhibitor.
10110. The method of item 10039 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
10111. The method of item 10039 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10112. The method of item 10039 wherein the anti-fibrotic agent is an MIF inhibitor.
10113. The method of item 10039 wherein the anti-fibrotic agent is an MMP inhibitor.
10114. The method of item 10039 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10115. The method of item 10039 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
10116. The method of item 10039 wherein the anti-fibrotic agent is a nitric oxide agonist.
10117. The method of item 10039 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
10118. The method of item 10039 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof. 10119. The method of item 10039 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
10120. The method of item 10039 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10121. The method of item 10039 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4),
ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from
CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10122. The method of item 10039 wherein the anti-fibrotic agent is a phosphatase inhibitor.
10123. The method of item 10039 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6)
(Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
10124. The method of item 10039 wherein the anti-fibrotic agent is a PKC inhibitor.
10125. The method of item 10039 wherein the anti-fibrotic agent is a platelet activating factor antagonist. 10126. The method of item 10039 wherein the anti-fibrotic agent rived growth factor receptor kinase inhibitor.
10127. The method of item 10039 wherein the anti-fibrotic agent roxylase inhibitor.
10128. The method of item 10039 wherein the anti-fibrotic agent honuclear neutrophil inhibitor.
10129. The method of item 10039 wherein the anti-fibrotic agent ase B inhibitor.
10130. The method of item 10039.wherein the anti-fibrotic agent ase C stimulant.
10131. The method of item 10039 wherein the anti-fibrotic agent cleoside analogue.
10132. The method of item 10039 wherein the anti-fibrotic agent eptor P2X antagonist.
10133. The method of item 10039 wherein the anti-fibrotic agent e inhibitor.
10134. The method of item 10039 wherein the anti-fibrotic agent inhibitor of ErbB1 and ErbB2.
10135. The method of item 10039 wherein the anti-fibrotic agent side triphosphate reductase inhibitor.
10136. The method of item 10039 wherein the anti-fibrotic agent ntagonist.
10137. The method of item 10039 wherein the anti-fibrotic agent inhibitor. 10138. The method of item 10039 wherein the anti-fibrotic agent is an SRC inhibitor.
10139. The method of item 10039 wherein the anti-fibrotic agent is a stromelysin inhibitor.
10140. The method of item 10039 wherein the anti-fibrotic agent is an Syk kinase inhibitor.
10141. The method of item 10039 wherein the anti-fibrotic agent is a telomerase inhibitor.
10142. The method of item 10039 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS
No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
10143. The method of item 10039 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10144. The method of item 10039 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
10145. The method of item 10039 wherein the anti-fibrotic agent is a tubulin antagonist. 10146. The method of item 10039 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
10147. The method of item 10039 wherein the anti-fibrotic agent is a VEGF inhibitor.
10148. The method of item 10039 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
10149. The method of item 10039 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
10150. The method of item 10039 wherein the anti-fibrotic agent is AP-23573 (an mTOR inhibitor).
10151. The method of item 10039 wherein the anti-fibrotic agent is synthadotin (a tubulin antagonist).
10152. The method of item 10039 wherein the anti-fibrotic agent is S-0885 (a collagenase inhibitor).
10153. The method of item 10039 wherein the anti-fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
10154. The method of item 10039 wherein the anti-fibrotic agent is ixabepilone (an epithilone). 10155. The method of item 10039 wherein the anti-fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
10156. The method of item 10039 wherein the anti-fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
10157. The method of item 10039 wherein the anti-fibrotic agent is ABT-518 (an angiogenesis inhibitor).
10158. The method of item 10039 wherein the anti-fibrotic agent is combretastatin (an angiogenesis inhibitor).
10159. The method of item 10039 wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
10160. The method of item 10039 wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
10161. The method of item 10039 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
10162. The method of item 10039 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
10163. The method of item 10039 wherein the anti-infective agent is an anthracycline.
10164. The method of item 10039 wherein the anti-infective agent is doxorubicin.
10165. The method of item 10039 wherein the anti-infective agent is mitoxantrone.
10166. The method of item 10039 wherein the anti-infective agent is a fluoropyrimidine. 10167. The method of item 10039 wherein the anti-infective agent is 5-fluorouracil (5-FU).
10168. The method of item 10039 wherein the anti-infective agent is a folic acid antagonist.
10169. The method of item 10039 wherein the anti-infective agent is methotrexate.
10170. The method of item 10039 wherein the anti-infective agent is a podophylotoxin.
10171. The method of item 10039 wherein the anti-infective agent is etoposide.
10172. The method of item 10039 wherein the anti-infective agent is camptothecin.
10173. The method of item 10039 wherein the anti-infective agent is a hydroxyurea.
10174. The method of item 10039 wherein the anti-infective agent is a platinum complex.
10175. The method of item 10039 wherein the anti-infective agent is cisplatin.
10176. The method of item 10039 wherein the composition comprises an anti-thrombotic agent.
10177. The method of item 10039 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
10178. The method of item 10039 wherein the polymer is formed from reactants comprising protein. 10179. The method of item 10039 wherein the polymer is formed from reactants comprising carbohydrate.
10180. The method of item 10039 wherein the polymer is formed from reactants comprising biodegradable polymer.
10181. The method of item 10039 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
10182. The method of item 10039 wherein the polymer is formed from reactants comprising collagen.
10183. The method of item 10039 wherein the polymer is formed from reactants comprising methylated collagen.
10184. The method of item 10039 wherein the polymer is formed from reactants comprising fibrinogen.
10185. The method of item 10039 wherein the polymer is formed from reactants comprising thrombin.
10186. The method of item 10039 wherein the polymer is formed from reactants comprising blood plasma.
10187. The method of item 10039 wherein the polymer is formed from reactants comprising calcium salt.
10188. The method of item 10039 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
10189. The method of item 10039 wherein the polymer is formed from reactants comprising fibrinogen analog.
10190. The method of item 10039 wherein the polymer is formed from reactants comprising albumin. 10191. The method of item 10039 wherein the polymer is formed from reactants comprising plasminogen.
10192. The method of item 10039 wherein the polymer is formed from reactants comprising von Willebrands factor.
10193. The method of item 10039 wherein the polymer is formed from reactants comprising Factor VIII.
10194. The method of item 10039 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
10195. The method of item 10039 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
10196. The method of item 10039 wherein the polymer is formed from reactants comprising telopeptide collagen.
10197. The method of item 10039 wherein the polymer is formed from reactants comprising crosslinked collagen.
10198. The method of item 10039 wherein the polymer is formed from reactants comprising aprotinin.
10199. The method of item 10039 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
10200. The method of item 10039 wherein the polymer is formed from reactants comprising gelatin.
10201. The method of item 10039 wherein the polymer is formed from reactants comprising protein conjugates.
10202. The method of item 10039 wherein the polymer is formed i from reactants comprising gelatin conjugates. 10203. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic polymer.
10204. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
10205. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
10206. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
10207. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
10208. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
10209. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
10210. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
10211. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
10212. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 10213. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
10214. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
10215. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
10216. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
10217. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
10218. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
10219. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
10220. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
10221. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups. 10222. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
10223. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
10224. The method of item 10039 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
10225. The method of item 10039 wherein the polymer is formed from reactants comprising polylysine.
10226. The method of item 10039 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
10227. The method of item 10039 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
10228. The method of item 10039 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
10229. The method of item 10039 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
10230. The method of item 10039 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 10231. The method of item 10039 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10232. The method of item 10039 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
10233. The method of item 10039 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
10234. The method of item 10039 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
10235. The method of item 10039 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
10236. The method of item 10039 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10237. The method of item 10039 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10238. The method of item 10039 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
10239. The method of item 10039 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine. 10240. The method of item 10039 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
10241. The method of item 10039 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
10242. The method of item 10039 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10243. The method of item 10039 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10244. The method of item 10039 wherein the polymer is formed from reactants comprising hyaluronic acid.
10245. The method of item 10039 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
10246. The method of item 10039 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
10247. The method of item 10039 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
10248. The method of item 10039 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
10249. The method of item 10039 wherein the composition comprises a colorant.
10250. The method of item 10039 wherein the composition is sterile.
10251. The method of any one of items 10039-10250 wherein the device monitors blood pressure.
10252. The method of any one of items 10039-10250 wherein the device monitors fluid flow.
10253. The method of any one of items 10039-10250 wherein the device monitors pressure within an aneurysm sac.
10254. The method of any one of items 10039-10250 wherein the device monitors intracranial pressure.
10255. The method of any one of items 10039-10250 wherein the device monitors mechanical pressure associated with a bone fracture.
10256. The method of any one of items 10039-10250 wherein the device monitors barometric pressure.
10257. The method of any one of items 10039-10250 wherein the device monitors eye tremors.
10258. The method of any one of items 10039-10250 wherein the device monitors the depth of a corneal implant.
10259. The method of any one of items 10039-10250 wherein the device monitors intraocular pressure. 10260. The method of any one of items 10039-10250 wherein the device is a passive sensor with an inductor-capacitor circuit.
10261. The method of any one of items 10039-10250 wherein the device is a self-powered strain sensing system.
10262. The method of any one of items 10039-10250 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage.
10263. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a cardiac sensor.
10264. The method for implanting a medical device according to item 10263 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
10265. The method for implanting a medical device according to item 10263 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
10266. The method for implanting a medical device according to item 10263 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host. 10267. The method for implanting a medical device according to item 10263 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host.
10268. The method for implanting a medical device according to item 10263 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
10269. The method for implanting a medical device according to item 10263 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
10270. The method of item 10263 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
10271. The method of item 10263 wherein the anti-fibrotic agent is an AKT inhibitor.
10272. The method of item 10263 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10273. The method of item 10263 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
10274. The method of item 10263 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist. 10275. The method of item 10263 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB)1 JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 10276. The method of item 10263 wherein the anti-fibrotic agent s an apoptosis antagonist.
10277. The method of item 10263 wherein the anti-fibrotic agent is an apoptosis activator.
10278. The method of item 10263 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
10279. The method of item 10263 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
10280. The method of item 10263 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
10281. The method of item 10263 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
10282. The method of item 10263 wherein the anti-fibrotic agent is a calcineurin inhibitor.
10283. The method of item 10263 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
10284. The method of item 10263 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
10285. The method of item 10263 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595
(Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10286. The method of item 10263 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
10287. The method of item 10263 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10288. The method of item 10263 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
10289. The method of item 10263 wherein the anti-fibrotic agent is a CD40 antagonist.
10290. The method of item 10263 wherein the anti-fibrotic agent is a chemokine receptor agonist.
10291. The method of item 10263 wherein the anti-fibrotic agent is a chymase inhibitor.
10292. The method of item 10263 wherein the anti-fibrotic agent is a collagenase antagonist.
10293. The method of item 10263 wherein the anti-fibrotic agent is a CXCR antagonist.
10294. The method of item 10263 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
10295. The method of item 10263 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor. 10296. The method of item 10263 wherein the anti-fibrotic agent is a DHFR inhibitor.
10297. The method of item 10263 wherein the anti-fibrotic agent is a dual integrin inhibitor.
10298. The method of item 10263 wherein the anti-fibrotic agent is an elastase inhibitor.
10299. The method of item 10263 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
10300. The method of item 10263 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
10301. The method of item 10263 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SlM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10302. The method of item 10263 wherein the anti-fibrotic agent is an endotoxin antagonist.
10303. The method of item 10263 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
10304. The method of item 10263 wherein the anti-fibrotic agent is an estrogen receptor antagonist. 10305. The method of item 10263 wherein the anti-fibrotic agent is an FGF inhibitor.
10306. The method of item 10263 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor.
10307. The method of item 10263 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10308. The method of item 10263 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
10309. The method of item 10263 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
10310. The method of item 10263 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10311. The method of item 10263 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof. 11726
10312. The method of item 10263 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
10313. The method of item 10263 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10314. The method of item 10263 wherein the anti-fibrotic agent is an ICAM inhibitor.
10315. The method of item 10263 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10316. The method of item 10263 wherein the anti-fibrotic agent is an IL-2 inhibitor.
10317. The method of item 10263 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SG N-35 '(Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
10318. The method of item 10263 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
10319. The method of item 10263 wherein the anti-fibrotic agent is an integrin antagonist.
10320. The method of item 10263 wherein the anti-fibrotic agent is an interleukin antagonist.
10321. The method of item 10263 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
10322. The method of item 10263 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
10323. The method of item 10263 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
10324. The method of item 10263 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
10325. The method of item 10263 wherein the anti-fibrotic agent is a JNK inhibitor.
10326. The method of item 10263 wherein the anti-fibrotic agent is a kinase inhibitor.
10327. The method of item 10263 wherein the anti-fibrotic agent is kinesin antagonist. 10328. The method of item 10263 wherein the anti-fibrotic agent is a kinesin antagonist.
10329. The method of item 10263 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10330. The method of item 10263 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
10331. The method of item 10263 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
10332. The method of item 10263 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
10333. The method of item 10263 wherein the anti-fibrotic agent is an mTOR inhibitor.
10334. The method of item 10263 wherein the anti-fibrotic agent is an mTOR kinase inhibitor. 006/011726
10335. The method of item 10263 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10336. The method of item 10263 wherein the anti-fibrotic agent is an MIF inhibitor.
10337. The method of item 10263 wherein the anti-fibrotic agent is an MMP inhibitor.
10338. The method of item 10263 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10339. The method of item 10263 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
10340. The method of item 10263 wherein the anti-fibrotic agent is a nitric oxide agonist.
10341. The method of item 10263 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
10342. The method of item 10263 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10343. The method of item 10263 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
10344. The method of item 10263 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10345. The method of item 10263 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10346. The method of item 10263 wherein the anti-fibrotic agent is a phosphatase inhibitor.
10347. The method of item 10263 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
10348. The method of item 10263 wherein the anti-fibrotic agent is a PKC inhibitor.
10349. The method of item 10263 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
10350. The method of item 10263 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
10351. The method of item 10263 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
10352. The method of item 10263 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
10353. The method of item 10263 wherein the anti-fibrotic agent is a protein kinase B inhibitor. 10354. The method of item 10263 wherein the anti-fibrotic agent ase C stimulant.
10355. The method of item 10263 wherein the anti-fibrotic agent cleoside analogue.
10356. The method of item 10263 wherein the anti-fibrotic agent eptor P2X antagonist.
10357. The method of item 10263 wherein the anti-fibrotic agent e inhibitor.
10358. The method of item 10263 wherein the anti-fibrotic agent inhibitor of ErbB1 and ErbB2.
10359. The method of item 10263 wherein the anti-fibrotic agent side triphosphate reductase inhibitor.
10360. The method of item 10263 wherein the anti-fibrotic agent ntagonist.
10361. The method of item 10263 wherein the anti-fibrotic agent inhibitor.
10362. The method of item 10263 wherein the anti-fibrotic agent ibitor.
10363. The method of item 10263 wherein the anti-fibrotic agent in inhibitor.
10364. The method of item 10263 wherein the anti-fibrotic agent se inhibitor.
10365. The method of item 10263 wherein the anti-fibrotic agent e inhibitor. 10366. The method of item 10263 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
10367. The method of item 10263 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 6 011726
4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10368. The method of item 10263 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
10369. The method of item 10263 wherein the anti-fibrotic agent is a tubulin antagonist.
10370. The method of item 10263 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248,
SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 10371. The method of item 10263 wherein the anti-fibrotic agent ibitor.
10372. The method of item 10263 wherein the anti-fibrotic agent receptor agonist.
10373. The method of item 10263 wherein the anti-fibrotic agent n angiogenesis inhibitor).
10374. The method of item 10263 wherein the anti-fibrotic agent an mTOR inhibitor).
10375. The method of item 10263 wherein the anti-fibrotic agent (a tubulin antagonist).
10376. The method of item 10263 wherein the anti-fibrotic agent ollagenase inhibitor).
10377. The method of item 10263 wherein the anti-fibrotic agent elongation factor-1 alpha inhibitor).
10378. The method of item 10263 wherein the anti-fibrotic agent (an epithilone).
\ \
10379. The method of item 10263 wherein the anti-fibrotic agent an angiogenesis inhibitor).
10380. The method of item 10263 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
10381. The method of item 10263 wherein the anti-fibrotic agent n angiogenesis inhibitor).
10382. The method of item 10263 wherein the anti-fibrotic agent atin (an angiogenesis inhibitor). 10383. The method of item 10263 wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
10384. The method of item 10263 wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
10385. The method of item 10263 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
10386. The method of item 10263 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
10387. The method of item 10263 wherein the anti-infective agent is an anthracycline.
10388. The method of item 10263 wherein the anti-infective agent is doxorubicin.
10389. The method of item 10263 wherein the anti-infective agent is mitoxantrone.
10390. The method of item 10263 wherein the anti-infective agent is a fluoropyrimidine.
10391. The method of item 10263 wherein the anti-infective agent is 5-fluorouracil (5-FU).
10392. The method of item 10263 wherein the anti-infective agent is a folic acid antagonist.
10393. The method of item 10263 wherein the anti-infective agent is methotrexate.
10394. The method of item 10263 wherein the anti-infective agent is a podophylotoxin. 10395. The method of item 10263 wherein the anti-infective agent is etoposide.
10396. The method of item 10263 wherein the anti-infective agent is camptothecin.
10397. The method of item 10263 wherein the anti-infective agent is a hydroxyurea.
10398. The method of item 10263 wherein the anti-infective agent is a platinum complex.
10399. The method of item 10263 wherein the anti-infective agent is cisplatin.
10400. The method of item 10263 wherein the composition comprises an anti-thrombotic agent.
10401. The method of item 10263 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
10402. The method of item 10263 wherein the polymer is formed from reactants comprising protein.
10403. The method of item 10263 wherein the polymer is formed from reactants comprising carbohydrate.
10404. The method of item 10263 wherein the polymer is formed from reactants comprising biodegradable polymer.
10405. The method of item 10263 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
10406. The method of item 10263 wherein the polymer is formed from reactants comprising collagen. 10407. The method of item 10263 wherein the polymer is formed from reactants comprising methylated collagen.
10408. The method of item 10263 wherein the polymer is formed from reactants comprising fibrinogen.
10409. The method of item 10263 wherein the polymer is formed from reactants comprising thrombin.
10410. The method of item 10263 wherein the polymer is formed from reactants comprising blood plasma.
10411. The method of item 10263 wherein the polymer is formed from reactants comprising calcium salt.
10412. The method of item 10263 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
10413. The method of item 10263 wherein the polymer is formed from reactants comprising fibrinogen analog.
10414. The method of item 10263 wherein the polymer is formed from reactants comprising albumin.
10415. The method of item 10263 wherein the polymer is formed from reactants comprising plasminogen.
10416. The method of item 10263 wherein the polymer is formed from reactants comprising von Willebrands factor.
10417. The method of item 10263 wherein the polymer is formed from reactants comprising Factor VIII.
10418. The method of item 10263 wherein the polymer is formed from reactants comprising hypoallergenic collagen. 10419. The method of item 10263 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
10420. The method of item 10263 wherein the polymer is formed from reactants comprising telopeptide collagen.
10421. The method of item 10263 wherein the polymer is formed from reactants comprising crosslinked collagen.
10422. The method of item 10263 wherein the polymer is formed from reactants comprising aprotinin.
10423. The method of item 10263 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
10424. The method of item 10263 wherein the polymer is formed from reactants comprising gelatin.
10425. The method of item 10263 wherein the polymer is formed from reactants comprising protein conjugates.
10426. The method of item 10263 wherein the polymer is formed from reactants comprising gelatin conjugates.
10427. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic polymer.
10428. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
10429. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
10430. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups. 10431. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
10432. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
10433. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
10434. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
10435. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
10436. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
10437. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
10438. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
10439. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups. 10440. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
10441. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
10442. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
10443. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
10444. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
10445. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
10446. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
10447. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
10448. The method of item 10263 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 10449. The method of item 10263 wherein the polymer is formed from reactants comprising polylysine.
10450. The method of item 10263 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
10451. The method of item 10263 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
10452. The method of item 10263 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
10453. The method of item 10263 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
10454. The method of item 10263 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10455. The method of item 10263 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10456. The method of item 10263 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
10457. The method of item 10263 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 10458. The method of item 10263 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
10459. The method of item 10263 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
10460. The method of item 10263 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10461. The method of item 10263 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10462. The method of item 10263 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
10463. The method of item 10263 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
10464. The method of item 10263 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
10465. The method of item 10263 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
10466. The method of item 10263 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 10467. The method of item 10263 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10468. The method of item 10263 wherein the polymer is formed from reactants comprising hyaluronic acid.
10469. The method of item 10263 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
10470. The method of item 10263 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
10471. The method of item 10263 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
10472. The method of item 10263 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
10473. The method of item 10263 wherein the composition comprises a colorant.
10474. The method of item 10263 wherein the composition is sterile.
10475. The method of any one of items 10263-10474 wherein the device monitors cardiac output. 10476. The method of any one of items 10263-10474 wherein the device monitors ejection fraction.
10477. The method of any one of items 10263-10474 wherein the device monitors blood pressure in a heart chamber.
10478. The method of any one of items 10263-10474 wherein the device monitors ventricular wall motions.
10479. The method of any one of items 10263-10474 wherein the device monitors blood flow to a transplanted organ.
10480. The method of any one of items 10263-10474 wherein the device monitors heart rate.
10481. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a respiratory sensor.
10482. The method for implanting a medical device according to item 10481 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
10483. The method for implanting a medical device according to item 10481 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 10484. The method for implanting a medical device according to item 10481 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
10485. The method for implanting a medical device according to item 10481 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host.
10486. The method for implanting a medical device according to item 10481 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
10487. The method for implanting a medical device according to item 10481 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
10488. The method of item 10481 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
10489. The method of item 10481 wherein the anti-fibrotic agent is an AKT inhibitor.
10490. The method of item 10481 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is
Pharmaprojects No. 5754 (Merck KgaA). 10491. The method of item 10481 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
10492. The method of item 10481 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
10493. The method of item 10481 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458
(Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedϊx), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10494. The method of item 10481 wherein the anti-fibrotic agent is an apoptosis antagonist.
10495. The method of item 10481 wherein the anti-fibrotic agent is an apoptosis activator.
10496. The method of item 10481 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
10497. The method of item 10481 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
10498. The method of item 10481 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
10499. The method of item 10481 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
10500. The method of item 10481 wherein the anti-fibrotic agent is a calcineurin inhibitor.
10501. The method of item 10481 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
10502. The method of item 10481 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
10503. The method of item 10481 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 10504. The method of item 10481 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
10505. The method of item 10481 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10506. The method of item 10481 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
10507. The method of item 10481 wherein the anti-fibrotic agent is a CD40 antagonist.
10508. The method of item 10481 wherein the anti-fibrotic agent is a chemokine receptor agonist.
10509. The method of item 10481 wherein the anti-fibrotic agent is a chymase inhibitor.
10510. The method of item 10481 wherein the anti-fibrotic agent is a collagenase antagonist.
10511. The method of item 10481 wherein the anti-fibrotic agent is a CXCR antagonist.
10512. The method of item 10481 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a
CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
10513. The method of item 10481 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
10514. The method of item 10481 wherein the anti-fibrotic agent is a DHFR inhibitor.
10515. The method of item 10481 wherein the anti-fibrotic agent is a dual integrin inhibitor.
10516. The method of item 10481 wherein the anti-fibrotic agent is an elastase inhibitor.
10517. The method of item 10481 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
10518. The method of item 10481 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
10519. The method of item 10481 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10520. The method of item 10481 wherein the anti-fibrotic agent is an endotoxin antagonist. 10521. The method of item 10481 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
10522. The method of item 10481 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
10523. The method of item 10481 wherein the anti-fibrotic agent is an FGF inhibitor.
10524. The method of item 10481 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor.
10525. The method of item 10481 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10526. The method of item 10481 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
10527. The method of item 10481 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
10528. The method of item 10481 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201
(Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10529. The method of item 10481 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG)1 rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
10530. The method of item 10481 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
10531. The method of item 10481 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10532. The method of item 10481 wherein the anti-fibrotic agent is an ICAM inhibitor.
10533. The method of item 10481 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-
424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10534. The method of item 10481 wherein the anti-fibrotic agent is an IL-2 inhibitor.
10535. The method of item 10481 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide
(Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protaiex), and an analogue or derivative thereof.
10536. The method of item 10481 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
10537. The method of item 10481 wherein the anti-fibrotic agent is an integrin antagonist.
10538. The method of item 10481 wherein the anti-fibrotic agent is an interleukin antagonist.
10539. The method of item 10481 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
10540. The method of item 10481 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
10541. The method of item 10481 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
10542. The method of item 10481 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
10543. The method of item 10481 wherein the anti-fibrotic agent is a JNK inhibitor.
10544. The method of item 10481 wherein the anti-fibrotic agent is a kinase inhibitor. 10545. The method of item 10481 wherein the anti-fibrotic agent is kinesin antagonist.
10546. The method of item 10481 wherein the anti-fibrotic agent is a kinesin antagonist.
10547. The method of item 10481 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingeiheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10548. The method of item 10481 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
10549. The method of item 10481 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
10550. The method of item 10481 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
10551. The method of item 10481 wherein the anti-fibrotic agent is an mTOR inhibitor. 10552. The method of item 10481 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
10553. The method of item 10481 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10554. The method of item 10481 wherein the anti-fibrotic agent is an MIF inhibitor.
10555. The method of item 10481 wherein the anti-fibrotic agent is an MMP inhibitor.
10556. The method of item 10481 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457
(Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 10557. The method of item 10481 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
10558. The method of item 10481 wherein the anti-fibrotic agent is a nitric oxide agonist.
10559. The method of item 10481 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
10560. The method of item 10481 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10561. The method of item 10481 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
10562. The method of item 10481 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-
993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10563. The method of item 10481 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735
(GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus
Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0)
(GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10564. The method of item 10481 wherein the anti-fibrotic agent is a phosphatase inhibitor.
10565. The method of item 10481 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
10566. The method of item 10481 wherein the anti-fibrotic agent is a PKC inhibitor.
10567. The method of item 10481 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
10568. The method of item 10481 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
10569. The method of item 10481 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
10570. The method of item 10481 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor. 10571. The method of item 10481 wherein the anti-fibrotic agent ase B inhibitor.
10572. The method of item 10481 wherein the anti-fibrotic agent ase C stimulant.
10573. The method of item 10481 wherein the anti-fibrotic agent cleoside analogue.
10574. The method of item 10481 wherein the anti-fibrotic agent eptor P2X antagonist.
10575. The method of item 10481 wherein the anti-fibrotic agent e inhibitor.
10576. The method of item 10481 wherein the anti-fibrotic agent inhibitor of ErbB1 and ErbB2.
10577. The method of item 10481 wherein the anti-fibrotic agent side triphosphate reductase inhibitor.
10578. The method of item 10481 wherein the anti-fibrotic agent ntagonist.
10579. The method of item 10481 wherein the anti-fibrotic agent inhibitor.
10580. The method of item 10481 wherein the anti-fibrotic agent ibitor.
10581. The method of item 10481 wherein the anti-fibrotic agent in inhibitor.
10582. The method of item 10481 wherein the anti-fibrotic agent se inhibitor. 10583. The method of item 10481 wherein the anti-fibrotic agent is a telomerase inhibitor.
10584. The method of item 10481 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
10585. The method of item 10481 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10586. The method of item 10481 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
10587. The method of item 10481 wherein the anti-fibrotic agent is a tubulin antagonist.
10588. The method of item 10481 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHlR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG)1 an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
10589. The method of item 10481 wherein the anti-fibrotic agent is a VEGF inhibitor.
10590. The method of item 10481 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
10591. The method of item 10481 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
10592. The method of item 10481 wherein the anti-fibrotic agent is AP-23573 (an mTOR inhibitor).
10593. The method of item 10481 wherein the anti-fibrotic agent is synthadotin (a tubulin antagonist).
10594. The method of item 10481 wherein the anti-fibrotic agent is S-0885 (a collagenase inhibitor).
10595. The method of item 10481 wherein the anti-fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
10596. The method of item 10481 wherein the anti-fibrotic agent is ixabepilone (an epithilone).
10597. The method of item 10481 wherein the anti-fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
10598. The method of item 10481 wherein the anti-fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
10599. The method of item 10481 wherein the anti-fibrotic agent is ABT-518 (an angiogenesis inhibitor). 10600. The method of item 10481 wherein the anti-fibrotic agent is combretastatin (an angiogenesis inhibitor).
10601. The method of item 10481 wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
10602. The method of item 10481 wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
10603. The method of item 10481 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
10604. The method of item 10481 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
10605. The method of item 10481 wherein the anti-infective agent is an anthracycline.
10606. The method of item 10481 wherein the anti-infective agent is doxorubicin.
10607. The method of item 10481 wherein the anti-infective agent is mitoxantrone.
10608. The method of item 10481 wherein the anti-infective agent is a fluoropyrimidine.
10609. The method of item 10481 wherein the anti-infective agent is 5-fluorouracil (5-FU).
10610. The method of item 10481 wherein the anti-infective agent is a folic acid antagonist.
10611. The method of item 10481 wherein the anti-infective agent is methotrexate. 10612. The method of item 10481 wherein the anti-infective agent is a podophylotoxin.
10613. The method of item 10481 wherein the anti-infective agent is etoposide.
10614. The method of item 10481 wherein the anti-infective agent is camptothecin.
10615. The method of item 10481 wherein the anti-infective agent is a hydroxyurea.
10616. The method of item 10481 wherein the anti-infective agent is a platinum complex.
10617. The method of item 10481 wherein the anti-infective agent is cisplatin.
10618. The method of item 10481 wherein the composition comprises an anti-thrombotic agent.
10619. The method of item 10481 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
10620. The method of item 10481 wherein the polymer is formed from reactants comprising protein.
10621. The method of item 10481 wherein the polymer is formed from reactants comprising carbohydrate.
10622. The method of item 10481 wherein the polymer is formed from reactants comprising biodegradable polymer.
10623. The method of item 10481 wherein the polymer is formed from reactants comprising nonbiodegradable polymer. 10624. The method of item 10481 wherein the polymer is formed from reactants comprising collagen.
10625. The method of item 10481 wherein the polymer is formed from reactants comprising methylated collagen.
10626. The method of item 10481 wherein the polymer is formed from reactants comprising fibrinogen.
10627. The method of item 10481 wherein the polymer is formed from reactants comprising thrombin.
10628. The method of item 10481 wherein the polymer is formed from reactants comprising blood plasma.
10629. The method of item 10481 wherein the polymer is formed from reactants comprising calcium salt.
10630. The method of item 10481 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
10631. The method of item 10481 wherein the polymer is formed from reactants comprising fibrinogen analog.
10632. The method of item 10481 wherein the polymer is formed from reactants comprising albumin.
10633. The method of item 10481 wherein the polymer is formed from reactants comprising plasminogen.
10634. The method of item 10481 wherein the polymer is formed from reactants comprising von Willebrands factor.
10635. The method of item 10481 wherein the polymer is formed from reactants comprising Factor VIII. 10636. The method of item 10481 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
10637. The method of item 10481 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
10638. The method of item 10481 wherein the polymer is formed from reactants comprising telopeptide collagen.
10639. The method of item 10481 wherein the polymer is formed from reactants comprising crosslinked collagen.
10640. The method of item 10481 wherein the polymer is formed from reactants comprising aprotinin.
10641. The method of item 10481 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
10642. The method of item 10481 wherein the polymer is formed from reactants comprising gelatin.
10643. The method of item 10481 wherein the polymer is formed from reactants comprising protein conjugates.
10644. The method of item 10481 wherein the polymer is formed from reactants comprising gelatin conjugates.
10645. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic polymer.
10646. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
10647. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 10648. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
10649. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
10650. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
10651. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
10652. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
10653. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
10654. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
10655. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
10656. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 10657. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three ' carbonyl-oxygen-succinimidyl groups.
10658. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
10659. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
10660. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
10661. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
10662. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
10663. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
10664. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
10665. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide. 10666. The method of item 10481 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
10667. The method of item 10481 wherein the polymer is formed from reactants comprising polylysine.
10668. The method of item 10481 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
10669. The method of item 10481 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
10670. The method of item 10481 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
10671. The method of item 10481 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
10672. The method of item 10481 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10673. The method of item 10481 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10674. The method of item 10481 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 10675. The method of item 10481 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
10676. The method of item 10481 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
10677. The method of item 10481 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
10678. The method of item 10481 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10679. The method of item 10481 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10680. The method of item 10481 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
10681. The method of item 10481 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
10682. The method of item 10481 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
10683. The method of item 10481 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups. 10684. The method of item 10481 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10685. The method of item 10481 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10686. The method of item 10481 wherein the polymer is formed from reactants comprising hyaluronic acid.
10687. The method of item 10481 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
10688. The method of item 10481 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
10689. The method of item 10481 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
10690. The method of item 10481 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
10691. The method of item 10481 wherein the composition comprises a colorant.
10692. The method of item 10481 wherein the composition is sterile. 10693. The method of any one of items 10481-10692 wherein the device monitors pulmonary functions.
10694. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an auditory sensor.
10695. The method for implanting a medical device according to item 10694 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
10696. The method for implanting a medical device according to item 10694 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
10697. The method for implanting a medical device according to item 10694 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
10698. The method for implanting a medical device according to item 10694 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host. 10699. The method for implanting a medical device according to item 10694 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
10700. The method for implanting a medical device according to item 10694 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
10701. The method of item 10694 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
10702. The method of item 10694 wherein the anti-fibrotic agent is an AKT inhibitor.
10703. The method of item 10694 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10704. The method of item 10694 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
10705. The method of item 10694 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
10706. The method of item 10694 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC)1 neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10707. The method of item 10694 wherein the anti-fibrotic agent is an apoptosis antagonist.
10708. The method of item 10694 wherein the anti-fibrotic agent is an apoptosis activator. 10709. The method of item 10694 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
10710. The method of item 10694 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
10711. The method of item 10694 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
10712. The method of item 10694 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
10713. The method of item 10694 wherein the anti-fibrotic agent is a calcineurin inhibitor.
10714. The method of item 10694 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
10715. The method of item 10694 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
10716. The method of item 10694 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10717. The method of item 10694 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
10718. The method of item 10694 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10719. The method of item 10694 wherein the anti-fibrotic agent is a cathepsin L inhibitor. 10720. The method of item 10694 wherein the anti-fibrotic agent is a CD40 antagonist.
10721. The method of item 10694 wherein the anti-fibrotic agent is a chemokine receptor agonist.
10722. The method of item 10694 wherein the anti-fibrotic agent is a chymase inhibitor.
10723. The method of item 10694 wherein the anti-fibrotic agent is a collagenase antagonist.
10724. The method of item 10694 wherein the anti-fibrotic agent is a CXCR antagonist.
10725. The method of item 10694 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
10726. The method of item 10694 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
10727. The method of item 10694 wherein the anti-fibrotic agent is a DHFR inhibitor.
10728. The method of item 10694 wherein the anti-fibrotic agent is a dual integrin inhibitor. 10729. The method of item 10694 wherein the anti-fibrotic agent is an eiastase inhibitor.
10730. The method of item 10694 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
10731. The method of item 10694 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
10732. The method of item 10694 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10733. The method of item 10694 wherein the anti-fibrotic agent is an endotoxin antagonist.
10734. The method of item 10694 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
10735. The method of item 10694 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
10736. The method of item 10694 wherein the anti-fibrotic agent is an FGF inhibitor.
10737. The method of item 10694 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor. 10738. The method of item 10694 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10739. The method of item 10694 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
10740. The method of item 10694 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
10741. The method of item 10694 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10742. The method of item 10694 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methy!propyl)-1-oxo-), and an analogue or derivative thereof.
10743. The method of item 10694 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
10744. The method of item 10694 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10745. The method of item 10694 wherein the anti-fibrotic agent is an ICAM inhibitor.
10746. The method of item 10694 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10747. The method of item 10694 wherein the anti-fibrotic agent is an IL-2 inhibitor.
10748. The method of item 10694 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 10749. The method of item 10694 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
10750. The method of item 10694 wherein the anti-fibrotic agent is an integrin antagonist.
10751. The method of item 10694 wherein the anti-fibrotic agent is an interleukin antagonist.
10752. The method of item 10694 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
10753. The method of item 10694 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
10754. The method of item 10694 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
10755. The method of item 10694 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
10756. The method of item 10694 wherein the anti-fibrotic agent is a JNK inhibitor.
10757. The method of item 10694 wherein the anti-fibrotic agent is a kinase inhibitor.
10758. The method of item 10694 wherein the anti-fibrotic agent is kinesin antagonist.
10759. The method of item 10694 wherein the anti-fibrotic agent is a kinesin antagonist.
10760. The method of item 10694 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10761. The method of item 10694 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
10762. The method of item 10694 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
10763. The method of item 10694 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
10764. The method of item 10694 wherein the anti-fibrotic agent is an mTOR inhibitor.
10765. The method of item 10694 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
10766. The method of item 10694 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-
maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10767. The method of item 10694 wherein the anti-fibrotic agent is an MIF inhibitor.
10768. The method of item 10694 wherein the anti-fibrotic agent is an MMP inhibitor.
10769. The method of item 10694 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10770. The method of item 10694 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
10771. The method of item 10694 wherein the anti-fibrotic agent is a nitric oxide agonist.
10772. The method of item 10694 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
10773. The method of item 10694 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10774. The method of item 10694 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
10775. The method of item 10694 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10776. The method of item 10694 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4),
ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from
CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10777. The method of item 10694 wherein the anti-fibrotic agent is a phosphatase inhibitor.
10778. The method of item 10694 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
10779. The method of item 10694 wherein the anti-fibrotic agent is a PKC inhibitor.
10780. The method of item 10694 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
10781. The method of item 10694 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
10782. The method of item 10694 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
10783. The method of item 10694 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
10784. The method of item 10694 wherein the anti-fibrotic agent is a protein kinase B inhibitor.
10785. The method of item 10694 wherein the anti-fibrotic agent is a protein kinase C stimulant.
10786. The method of item 10694 wherein the anti-fibrotic agent is a purine nucleoside analogue.
10787. The method of item 10694 wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist. 10788. The method of item 10694 wherein the anti-fibrotic agent is a Raf kinase inhibitor.
10789. The method of item 10694 wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
10790. The method of item 10694 wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
10791. The method of item 10694 wherein the anti-fibrotic agent is an SDF-1 antagonist.
10792. The method of item 10694 wherein the anti-fibrotic agent is a sheddase inhibitor.
10793. The method of item 10694 wherein the anti-fibrotic agent is an SRC inhibitor.
10794. The method of item 10694 wherein the anti-fibrotic agent is a stromelysin inhibitor.
10795. The method of item 10694 wherein the anti-fibrotic agent is an Syk kinase inhibitor.
10796. The method of item 10694 wherein the anti-fibrotic agent is a teiomerase inhibitor.
10797. The method of item 10694 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS
No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 10798. The method of item 10694 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPFM 32294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10799. The method of item 10694 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
10800. The method of item 10694 wherein the anti-fibrotic agent is a tubulin antagonist.
10801. The method of item 10694 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
10802. The method of item 10694 wherein the anti-fibrotic agent is a VEGF inhibitor.
10803. The method of item 10694 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
10804. The method of item 10694 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor). 10805. The method of item 10694 wherein the anti-fibrotic agent an mTOR inhibitor).
10806. The method of item 10694 wherein the anti-fibrotic agent (a tubulin antagonist).
10807. The method of item 10694 wherein the anti-fibrotic agent ollagenase inhibitor).
10808. The method of item 10694 wherein the anti-fibrotic agent elongation factor-1 alpha inhibitor).
10809. The method of item 10694 wherein the anti-fibrotic agent (an epithilone).
10810. The method of item 10694 wherein the anti-fibrotic agent n angiogenesis inhibitor).
10811. The method of item 10694 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
10812. The method of item 10694 wherein the anti-fibrotic agent n angiogenesis inhibitor).
10813. The method of item 10694 wherein the anti-fibrotic agent tin (an angiogenesis inhibitor).
10814. The method of item 10694 wherein the anti-fibrotic agent acetate (an angiogenesis inhibitor).
10815. The method of item 10694 wherein the anti-fibrotic agent (a kinesin antagonist).
10816. The method of item 10694 wherein the anti-fibrotic agent s (an mTOR inhibitor). 10817. The method of item 10694 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
10818. The method of item 10694 wherein the anti-infective agent is an anthracycline.
10819. The method of item 10694 wherein the anti-infective agent is doxorubicin.
10820. The method of item 10694 wherein the anti-infective agent is mitoxantrone.
10821. The method of item 10694 wherein the anti-infective agent is a fluoropyrimidine.
10822. The method of item 10694 wherein the anti-infective agent is 5-fluorouracil (5-FU).
10823. The method of item 10694 wherein the anti-infective agent is a folic acid antagonist.
10824. The method of item 10694 wherein the anti-infective agent is methotrexate.
10825. The method of item 10694 wherein the anti-infective agent is a podophylotoxin.
10826. The method of item 10694 wherein the anti-infective agent is etoposide.
10827. The method of item 10694 wherein the anti-infective agent is camptothecin.
10828. The method of item 10694 wherein the anti-infective agent is a hydroxyurea. 10829. The method of item 10694 wherein the anti-infective agent is a platinum complex.
10830. The method of item 10694 wherein the anti-infective agent is cisplatin.
10831. The method of item 10694 wherein the composition comprises an anti-thrombotic agent.
10832. The method of item 10694 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
10833. The method of item 10694 wherein the polymer is formed from reactants comprising protein.
10834. The method of item 10694 wherein the polymer is formed from reactants comprising carbohydrate.
10835. The method of item 10694 wherein the polymer is formed from reactants comprising biodegradable polymer.
10836. The method of item 10694 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
10837. The method of item 10694 wherein the polymer is formed from reactants comprising collagen.
10838. The method of item 10694 wherein the polymer is formed from reactants comprising methylated collagen.
10839. The method of item 10694 wherein the polymer is formed from reactants comprising fibrinogen.
10840. The method of item 10694 wherein the polymer is formed from reactants comprising thrombin. 10841. The method of item 10694 wherein the polymer is formed from reactants comprising blood plasma.
10842. The method of item 10694 wherein the polymer is formed from reactants comprising calcium salt.
10843. The method of item 10694 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
10844. The method of item 10694 wherein the polymer is formed from reactants comprising fibrinogen analog.
10845. The method of item 10694 wherein the polymer is formed from reactants comprising albumin.
10846. The method of item 10694 wherein the polymer is formed from reactants comprising plasminogen.
10847. The method of item 10694 wherein the polymer is formed from reactants comprising von Willebrands factor.
10848. The method of item 10694 wherein the polymer is formed from reactants comprising Factor VIII.
10849. The method of item 10694 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
10850. The method of item 10694 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
10851. The method of item 10694 wherein the polymer is formed from reactants comprising telopeptide collagen.
10852. The method of item 10694 wherein the polymer is formed from reactants comprising crosslinked collagen. 10853. The method of item 10694 wherein the polymer is formed from reactants comprising aprotinin.
10854. The method of item 10694 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
10855. The method of item 10694 wherein the polymer is formed from reactants comprising gelatin.
10856. The method of item 10694 wherein the polymer is formed from reactants comprising protein conjugates.
10857. The method of item 10694 wherein the polymer is formed from reactants comprising gelatin conjugates.
10858. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic polymer.
10859. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
10860. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
10861. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
10862. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
10863. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 10864. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
10865. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
10866. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
10867. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
10868. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
10869. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
10870. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
10871. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
10872. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 10873. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
10874. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
10875. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
10876. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
10877. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
10878. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
10879. The method of item 10694 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
10880. The method of item 10694 wherein the polymer is formed from reactants comprising polylysine.
10881. The method of item 10694 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion. 10882. The method of item 10694 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
10883. The method of item 10694 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
10884. The method of item 10694 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
10885. The method of item 10694 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10886. The method of item 10694 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10887. The method of item 10694 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
10888. The method of item 10694 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
10889. The method of item 10694 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
10890. The method of item 10694 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups. 10891. The method of item 10694 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10892. The method of item 10694 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10893. The method of item 10694 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
10894. The method of item 10694 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
10895. The method of item 10694 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
10896. The method of item 10694 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
10897. The method of item 10694 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
10898. The method of item 10694 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
10899. The method of item 10694 wherein the polymer is formed from reactants comprising hyaluronic acid. 10900. The method of item 10694 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
10901. The method of item 10694 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
10902. The method of item 10694 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
10903. The method of item 10694 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
10904. The method of item 10694 wherein the composition comprises a colorant.
10905. The method of item 10694 wherein the composition is sterile.
10906. The method of any one of items 10694-10905 wherein the device is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
10907. The method of any one of items 10694-10905 wherein the device generates an electrical audio signal.
10908. The method of any one of items 10694-10905 wherein the device is a capacitive sensor that is coupled to a vibrating auditory element. 10909. The method of any one of items 10694-10905 wherein the device is an electromagnetic sensor.
10910. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an electrolyte or metabolite sensor.
10911. The method for implanting a medical device according to item 10910 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
10912. The method for implanting a medical device according to item 10910 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
10913. The method for implanting a medical device according to item 10910 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
10914. The method for implanting a medical device according to item 10910 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host. 10915. The method for implanting a medical device according to item 10910 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
10916. The method for implanting a medical device according to item 10910 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
10917. The method of item 10910 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
10918. The method of item 10910 wherein the anti-fibrotic agent is an AKT inhibitor.
10919. The method of item 10910 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10920. The method of item 10910 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
10921. The method of item 10910 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
10922. The method of item 10910 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), lDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherϊn (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10923. The method of item 10910 wherein the anti-fibrotic agent is an apoptosis antagonist.
10924. The method of item 10910 wherein the anti-fibrotic agent is an apoptosis activator. 10925. The method of item 10910 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
10926. The method of item 10910 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
10927. The method of item 10910 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
10928. The method of item 10910 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
10929. The method of item 10910 wherein the anti-fibrotic agent is a calcineurin inhibitor.
10930. The method of item 10910 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
10931. The method of item 10910 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
10932. The method of item 10910 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10933. The method of item 10910 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
10934. The method of item 10910 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10935. The method of item 10910 wherein the anti-fibrotic agent is a cathepsin L inhibitor. 10936. The method of item 10910 wherein the anti-fibrotic agent is a CD40 antagonist.
10937. The method of item 10910 wherein the anti-fibrotic agent is a chemokine receptor agonist.
10938. The method of item 10910 wherein the anti-fibrotic agent is a chymase inhibitor.
10939. The method of item 10910 wherein the anti-fibrotic agent is a collagenase antagonist.
10940. The method of item 10910 wherein the anti-fibrotic agent is a CXCR antagonist.
10941. The method of item 10910 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
10942. The method of item 10910 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
10943. The method of item 10910 wherein the anti-fibrotic agent is a DHFR inhibitor.
10944. The method of item 10910 wherein the anti-fibrotic agent is a dual integrin inhibitor. 10945. The method of item 10910 wherein the anti-fibrotic agent is an elastase inhibitor.
10946. The method of item 10910 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
10947. The method of item 10910 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
10948. The method of item 10910 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10949. The method of item 10910 wherein the anti-fibrotic agent is an endotoxin antagonist.
10950. The method of item 10910 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
10951. The method of item 10910 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
10952. The method of item 10910 wherein the anti-fibrotic agent is an FGF inhibitor.
10953. The method of item 10910 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor. 10954. The method of item 10910 wherein the anti-fibrotic agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10955. The method of item 10910 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
10956. The method of item 10910 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
10957. The method of item 10910 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB)1 plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10958. The method of item 10910 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy~1 ,4-dihydro-5'-(2~ methylpropyl)-1-oxo-), and an analogue or derivative thereof.
10959. The method of item 10910 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
10960. The method of item 10910 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10961. The method of item 10910 wherein the anti-fibrotic agent is an ICAM inhibitor.
10962. The method of item 10910 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10963. The method of item 10910 wherein the anti-fibrotic agent is an IL-2 inhibitor.
10964. The method of item 10910 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 10965. The method of item 10910 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
10966. The method of item 10910 wherein the anti-fibrotic agent is an integrin antagonist.
10967. The method of item 10910 wherein the anti-fibrotic agent is an interleukin antagonist.
10968. The method of item 10910 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
10969. The method of item 10910 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
10970. The method of item 10910 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
10971. The method of item 10910 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
10972. The method of item 10910 wherein the anti-fibrotic agent is a JNK inhibitor.
10973. The method of item 10910 wherein the anti-fibrotic agent is a kinase inhibitor.
10974. The method of item 10910 wherein the anti-fibrotic agent is kinesin antagonist.
10975. The method of item 10910 wherein the anti-fibrotic agent is a kinesin antagonist.
10976. The method of item 10910 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10977. The method of item 10910 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
10978. The method of item 10910 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
10979. The method of item 10910 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
10980. The method of item 10910 wherein the anti-fibrotic agent is an mTOR inhibitor.
10981. The method of item 10910 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
10982. The method of item 10910 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10983. The method of item 10910 wherein the anti-fibrotic agent is an MIF inhibitor.
10984. The method of item 10910 wherein the anti-fibrotic agent is an MMP inhibitor.
10985. The method of item 10910 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10986. The method of item 10910 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
10987. The method of item 10910 wherein the anti-fibrotic agent is a nitric oxide agonist.
10988. The method of item 10910 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
10989. The method of item 10910 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10990. The method of item 10910 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
10991. The method of item 10910 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10992. The method of item 10910 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- W
3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4),
ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from
CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10993. The method of item 10910 wherein the anti-fibrotic agent is a phosphatase inhibitor.
10994. The method of item 10910 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
10995. The method of item 10910 wherein the anti-fibrotic agent is a PKC inhibitor.
10996. The method of item 10910 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
10997. The method of item 10910 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
10998. The method of item 10910 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
10999. The method of item 10910 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
11000. The method of item 10910 wherein the anti-fibrotic agent is a protein kinase B inhibitor.
11001. The method of item 10910 wherein the anti-fibrotic agent is a protein kinase C stimulant.
11002. The method of item 10910 wherein the anti-fibrotic agent is a purine nucleoside analogue.
11003. The method of item 10910 wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist. 11004. The method of item 10910 wherein the anti-fibrotic agent is a Raf kinase inhibitor.
11005. The method of item 10910 wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
11006. The method of item 10910 wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
11007. The method of item 10910 wherein the anti-fibrotic agent is an SDF-1 antagonist.
11008. The method of item 10910 wherein the anti-fibrotic agent is a sheddase inhibitor.
11009. The method of item 10910 wherein the anti-fibrotic agent is an SRC inhibitor.
11010. The method of item 10910 wherein the anti-fibrotic agent is a stromelysin inhibitor.
11011. The method of item 10910 wherein the anti-fibrotic agent is an Syk kinase inhibitor.
11012. The method of item 10910 wherein the anti-fibrotic agent is a telomerase inhibitor.
11013. The method of item 10910 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS
No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 11014. The method of item 10910 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersaiazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11015. The method of item 10910 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
11016. The method of item 10910 wherein the anti-fibrotic agent is a tubulin antagonist.
11017. The method of item 10910 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
11018. The method of item 10910 wherein the anti-fibrotic agent is a VEGF inhibitor.
11019. The method of item 10910 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
11020. The method of item 10910 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor). 11021. The method of item 10910 wherein the anti-fibrotic agent an mTOR inhibitor).
11022. The method of item 10910 wherein the anti-fibrotic agent (a tubulin antagonist).
11023. The method of item 10910 wherein the anti-fibrotic agent ollagenase inhibitor).
11024. The method of item 10910 wherein the anti-fibrotic agent elongation factor-1 alpha inhibitor).
11025. The method of item 10910 wherein the anti-fibrotic agent (an epithilone).
11026. The method of item 10910 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11027. The method of item 10910 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
11028. The method of item 10910 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11029. The method of item 10910 wherein the anti-fibrotic agent atin (an angiogenesis inhibitor).
11030. The method of item 10910 wherein the anti-fibrotic agent acetate (an angiogenesis inhibitor).
11031. The method of item 10910 wherein the anti-fibrotic agent (a kinesin antagonist).
11032. The method of item 10910 wherein the anti-fibrotic agent s (an mTOR inhibitor). ( 11033. The method of item 10910 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
11034. The method of item 10910 wherein the anti-infective agent is an anthracycline.
11035. The method of item 10910 wherein the anti-infective agent is doxorubicin.
11036. The method of item 10910 wherein the anti-infective agent is mitoxantrone.
11037. The method of item 10910 wherein the anti-infective agent is a fluoropyrimidine.
11038. The method of item 10910 wherein the anti-infective agent is 5-fluorouracil (5-FU).
11039. The method of item 10910 wherein the anti-infective agent is a folic acid antagonist.
11040. The method of item 10910 wherein the anti-infective agent is methotrexate.
11041. The method of item 10910 wherein the anti-infective agent is a podophylotoxin.
11042. The method of item 10910 wherein the anti-infective agent is etoposide.
11043. The method of item 10910 wherein the anti-infective agent is camptothecin.
11044. The method of item 10910 wherein the anti-infective agent is a hydroxyurea. 11045. The method of item 10910 wherein the anti-infective agent is a platinum complex.
11046. The method of item 10910 wherein the anti-infective agent is cisplatϊn.
11047. The method of item 10910 wherein the composition comprises an anti-thrombotic agent.
11048. The method of item 10910 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
11049. The method of item 10910 wherein the polymer is formed from reactants comprising protein.
11050. The method of item 10910 wherein the polymer is formed from reactants comprising carbohydrate.
11051. The method of item 10910 wherein the polymer is formed from reactants comprising biodegradable polymer.
11052. The method of item 10910 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
11053. The method of item 10910 wherein the polymer is formed from reactants comprising collagen.
11054. The method of item 10910 wherein the polymer is formed from reactants comprising methylated collagen.
11055. The method of item 10910 wherein the polymer is formed from reactants comprising fibrinogen.
11056. The method of item 10910 wherein the polymer is formed from reactants comprising thrombin. 11057. The method of item 10910 wherein the polymer is formed from reactants comprising blood plasma.
11058. The method of item 10910 wherein the polymer is formed from reactants comprising calcium salt.
11059. The method of item 10910 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
11060. The method of item 10910 wherein the polymer is formed from reactants comprising fibrinogen analog.
11061. The method of item 10910 wherein the polymer is formed from reactants comprising albumin.
11062. The method of item 10910 wherein the polymer is formed from reactants comprising plasminogen.
11063. The method of item 10910 wherein the polymer is formed from reactants comprising von Willebrands factor.
11064. The method of item 10910 wherein the polymer is formed from reactants comprising Factor VIII.
11065. The method of item 10910 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
11066. The method of item 10910 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
11067. The method of item 10910 wherein the polymer is formed from reactants comprising telopeptide collagen.
11068. The method of item 10910 wherein the polymer is formed from reactants comprising crosslinked collagen. 11069. The method of item 10910 wherein the polymer is formed from reactants comprising aprotinin.
11070. The method of item 10910 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
11071. The method of item 10910 wherein the polymer is formed from reactants comprising gelatin.
11072. The method of item 10910 wherein the polymer is formed from reactants comprising protein conjugates.
11073. The method of item 10910 wherein the polymer is formed from reactants comprising gelatin conjugates.
11074. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic polymer.
11075. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
11076. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
11077. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
11078. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
11079. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 11080. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
11081. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
11082. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
11083. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
11084. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
11085. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
11086. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
11087. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
11088. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 11089. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
11090. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
11091. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
11092. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
11093. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
11094. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
11095. The method of item 10910 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
11096. The method of item 10910 wherein the polymer is formed from reactants comprising polylysine.
11097. The method of item 10910 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion. 11098. The method of item 10910 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
11099. The method of item 10910 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
11100. The method of item 10910 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
11101. The method of item 10910 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11102. The method of item 10910 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11103. The method of item 10910 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
11104. The method of item 10910 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
11105. The method of item 10910 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
11106. The method of item 10910 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups. 11107. The method of item 10910 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11108. The method of item 10910 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11109. The method of item 10910 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
11110. The method of item 10910 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
11111. The method of item 10910 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
11112. The method of item 10910 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
11113. The method of item 10910 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11114. The method of item 10910 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11115. The method of item 10910 wherein the polymer is formed from reactants comprising hyaluronic acid. 11116. The method of item 10910 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
11117. The method of item 10910 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
11118. The method of item 10910 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
11119. The method of item 10910 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
11120. The method of item 10910 wherein the composition comprises a colorant.
11121. The method of item 10910 wherein the composition is sterile.
11122. The method of any one of items 10910-11121 wherein the device emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
11123. The method of any one of items 10910-11121 wherein the device is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader. 11124. The method of any one of items 10910-11121 wherein the device is a monolithic bioelectronic device for detecting at least one analyte within the host.
11125. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a pump.
11126. The method for implanting a medical device according to item 11125 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
11127. The method for implanting a medical device according to item 11125 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
11128. The method for implanting a medical device according to item 11125 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
11129. The method for implanting a medical device according to item 11125 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host. 11130. The method for implanting a medical device according to item 11125 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
11131. The method for implanting a medical device according to item 11125 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
11132. The method of item 11125 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
11133. The method of item 11125 wherein the anti-fibrotic agent is an AKT inhibitor.
11134. The method of item 11125 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11135. The method of item 11125 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
11136. The method of item 11125 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
11137. The method of item 11125 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
11138. The method of item 11125 wherein the anti-fibrotic agent is an apoptosis antagonist.
11139. The method of item 11125 wherein the anti-fibrotic agent is an apoptosis activator. 11140. The method of item 11125 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
11141. The method of item 11125 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
11142. The method of item 11125 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
11143. The method of item 11125 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
11144. The method of item 11125 wherein the anti-fibrotic agent is a calcineurin inhibitor.
11145. The method of item 11125 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
11146. The method of item 11125 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
11147. The method of item 11125 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11148. The method of item 11125 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
11149. The method of item 11125 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
11150. The method of item 11125 wherein the anti-fibrotic agent is a cathepsin L inhibitor. 11151. The method of item 11125 wherein the anti-fibrotic agent is a CD40 antagonist.
11152. The method of item 11125 wherein the anti-fibrotic agent is a chemokine receptor agonist.
11153. The method of item 11125 wherein the anti-fibrotic agent is a chymase inhibitor.
11154. The method of item 11125 wherein the anti-fibrotic agent is a collagenase antagonist.
11155. The method of item 11125 wherein the anti-fibrotic agent is a CXCR antagonist.
11156. The method of item 11125 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
11157. The method of item 11125 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
11158. The method of item 11125 wherein the anti-fibrotic agent is a DHFR inhibitor.
11159. The method of item 11125 wherein the anti-fibrotic agent is a dual integrin inhibitor. 11160. The method of item 11125 wherein the anti-fibrotic agent is an elastase inhibitor.
11161. The method of item 11125 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
11162. The method of item 11125 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
11163. The method of item 11125 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
11164. The method of item 11125 wherein the anti-fibrotic agent is an endotoxin antagonist.
11165. The method of item 11125 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
11166. The method of item 11125 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
11167. The method of item 11125 wherein the anti-fibrotic agent is an FGF inhibitor.
11168. The method of item 11125 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor. 11169. The method of item 11125 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11170. The method of item 11125 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
11171. The method of item 11125 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
11172. The method of item 11125 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11173. The method of item 11125 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
11174. The method of item 11125 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
11175. The method of item 11125 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11176. The method of item 11125 wherein the anti-fibrotic agent is an ICAM inhibitor.
11177. The method of item 11125 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11178. The method of item 11125 wherein the anti-fibrotic agent is an IL-2 inhibitor.
11179. The method of item 11125 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Ph a rma projects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 11180. The method of item 11125 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
11181. The method of item 11125 wherein the anti-fibrotic agent is an integrin antagonist.
11182. The method of item 11125 wherein the anti-fibrotic agent is an interleukin antagonist.
11183. The method of item 11125 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
11184. The method of item 11125 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11185. The method of item 11125 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
11186. The method of item 11125 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
11187. The method of item 11125 wherein the anti-fibrotic agent is a JNK inhibitor.
11188. The method of item 11125 wherein the anti-fibrotic agent is a kinase inhibitor.
11189. The method of item 11125 wherein the anti-fibrotic agent is kinesin antagonist.
11190. The method of item 11125 wherein the anti-fibrotic agent is a kinesin antagonist.
11191. The method of item 11125 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11192. The method of item 11125 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
11193. The method of item 11125 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
11194. The method of item 11125 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
11195. The method of item 11125 wherein the anti-fibrotic agent is an mTOR inhibitor.
11196. The method of item 11125 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
11197. The method of item 11125 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
11198. The method of item 11125 wherein the anti-fibrotic agent is an MIF inhibitor.
11199. The method of item 11125 wherein the anti-fibrotic agent is an MMP inhibitor.
11200. The method of item 11125 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11201. The method of item 11125 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11202. The method of item 11125 wherein the anti-fibrotic agent is a nitric oxide agonist.
11203. The method of item 11125 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
11204. The method of item 11125 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11205. The method of item 11125 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
11206. The method of item 11125 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
11207. The method of item 11125 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4),
ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from
CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11208. The method of item 11125 wherein the anti-fibrotic agent is a phosphatase inhibitor.
11209. The method of item 11125 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
11210. The method of item 11125 wherein the anti-fibrotic agent is a PKC inhibitor.
11211. The method of item 11125 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
11212. The method of item 11125 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
11213. The method of item 11125 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
11214. The method of item 11125 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
11215. The method of item 11125 wherein the anti-fibrotic agent is a protein kinase B inhibitor.
11216. The method of item 11125 wherein the anti-fibrotic agent is a protein kinase C stimulant.
11217. The method of item 11125 wherein the anti-fibrotic agent is a purine nucleoside analogue.
11218. The method of item 11125 wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist. 11219. The method of item 11125 wherein the anti-f ibrotic agent is a Raf kinase inhibitor.
11220. The method of item 11125 wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
11221. The method of item 11125 wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
11222. The method of item 11125 wherein the anti-fibrotic agent is an SDF-1 antagonist.
11223. The method of item 11125 wherein the anti-fibrotic agent is a sheddase inhibitor.
11224. The method of item 11125 wherein the anti-fibrotic agent is an SRC inhibitor.
11225. The method of item 11125 wherein the anti-fibrotic agent is a stromelysin inhibitor.
11226. The method of item 11125 wherein the anti-fibrotic agent is an Syk kinase inhibitor.
11227. The method of item 11125 wherein the anti-fibrotic agent is a telomerase inhibitor.
11228. The method of item 11125 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS
No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 11229. The method of item 11125 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11230. The method of item 11125 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
11231. The method of item 11125 wherein the anti-fibrotic agent is a tubulin antagonist.
11232. The method of item 11125 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), Iapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
11233. The method of item 11125 wherein the anti-fibrotic agent is a VEGF inhibitor.
11234. The method of item 11125 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
11235. The method of item 11125 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor). 11236. The method of item 11125 wherein the anti-fibrotic agent an mTOR inhibitor).
11237. The method of item 11125 wherein the anti-fibrotic agent (a tubulin antagonist).
11238. The method of item 11125 wherein the anti-fibrotic agent ollagenase inhibitor).
11239. The method of item 11125 wherein the anti-fibrotic agent elongation factor-1 alpha inhibitor).
11240. The method of item 11125 wherein the anti-fibrotic agent (an epithilone).
11241. The method of item 11125 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11242. The method of item 11125 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
11243. The method of item 11125 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11244. The method of item 11125 wherein the anti-fibrotic agent tin (an angiogenesis inhibitor).
11245. The method of item 11125 wherein the anti-fibrotic agent acetate (an angiogenesis inhibitor).
11246. The method of item 11125 wherein the anti-fibrotic agent (a kinesin antagonist).
11247. The method of item 11125 wherein the anti-fibrotic agent s (an mTOR inhibitor). 11248. The method of item 11125 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
11249. The method of item 11125 wherein the anti-infective agent is an anthracycline.
11250. The method of item 11125 wherein the anti-infective agent is doxorubicin.
11251. The method of item 11125 wherein the anti-infective agent is mitoxantrone.
11252. The method of item 11125 wherein the anti-infective agent is a fluoropyrimidine.
11253. The method of item 11125 wherein the anti-infective agent is 5-fluorouracil (5-FU).
11254. The method of item 11125 wherein the anti-infective agent is a folic acid antagonist.
11255. The method of item 11125 wherein the anti-infective agent is methotrexate.
11256. The method of item 11125 wherein the anti-infective agent is a podophylotoxin.
11257. The method of item 11125 wherein the anti-infective agent is etoposide.
11258. The method of item 11125 wherein the anti-infective agent is camptothecin.
11259. The method of item 11125 wherein the anti-infective agent is a hydroxyurea. 11260. The method of item 11125 wherein the anti-infective agent is a platinum complex.
11261. The method of item 11125 wherein the anti-infective agent is cisplatin.
11262. The method of item 11125 wherein the composition comprises an anti-thrombotic agent.
11263. The method of item 11125 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
11264. The method of item 11125 wherein the polymer is formed from reactants comprising protein.
11265. The method of item 11125 wherein the polymer is formed from reactants comprising carbohydrate.
11266. The method of item 11125 wherein the polymer is formed from reactants comprising biodegradable polymer.
11267. The method of item 11125 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
11268. The method of item 11125 wherein the polymer is formed from reactants comprising collagen.
11269. The method of item 11125 wherein the polymer is formed from reactants comprising methylated collagen.
11270. The method of item 11125 wherein the polymer is formed from reactants comprising fibrinogen.
11271. The method of item 11125 wherein the polymer is formed from reactants comprising thrombin. 11272. The method of item 11125 wherein the polymer is formed from reactants comprising blood plasma.
11273. The method of item 11125 wherein the polymer is formed from reactants comprising calcium salt.
11274. The method of item 11125 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
11275. The method of item 11125 wherein the polymer is formed from reactants comprising fibrinogen analog.
11276. The method of item 11125 wherein the polymer is formed from reactants comprising albumin.
11277. The method of item 11125 wherein the polymer is formed from reactants comprising plasminogen.
11278. The method of item 11125 wherein the polymer is formed from reactants comprising von Willebrands factor.
11279. The method of item 11125 wherein the polymer is formed from reactants comprising Factor VIII.
11280. The method of item 11125 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
11281. The method of item 11125 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
11282. The method of item 11125 wherein the polymer is formed from reactants comprising telopeptide collagen.
11283. The method of item 11125 wherein the polymer is formed from reactants comprising crosslinked collagen. 11284. The method of item 11125 wherein the polymer is formed from reactants comprising aprotinin.
11285. The method of item 11125 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
11286. The method of item 11125 wherein the polymer is formed from reactants comprising gelatin.
11287. The method of item 11125 wherein the polymer is formed from reactants comprising protein conjugates.
11288. The method of item 11125 wherein the polymer is formed from reactants comprising gelatin conjugates.
11289. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic polymer.
11290. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
11291. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
11292. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
11293. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
11294. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 11295. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
11296. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
11297. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
11298. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
11299. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
11300. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
11301. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
11302. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
11303. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 11304. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
11305. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
11306. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
11307. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
11308. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
11309. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
11310. The method of item 11125 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
11311. The method of item 11125 wherein the polymer is formed from reactants comprising polylysine.
11312. The method of item 11125 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion. 11313. The method of item 11125 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
11314. The method of item 11125 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
11315. The method of item 11125 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
11316. The method of item 11125 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11317. The method of item 11125 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11318. The method of item 11125 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
11319. The method of item 11125 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
11320. The method of item 11125 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
11321. The method of item 11125 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups. 11322. The method of item 11125 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11323. The method of item 11125 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11324. The method of item 11125 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
11325. The method of item 11125 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
11326. The method of item 11125 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
11327. The method of item 11125 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
11328. The method of item 11125 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11329. The method of item 11125 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11330. The method of item 11125 wherein the polymer is formed from reactants comprising hyaluronic acid. 11331. The method of item 11125 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
11332. The method of item 11125 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
11333. The method of item 11125 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
11334. The method of item 11125 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
11335. The method of item 11125 wherein the composition comprises a colorant.
11336. The method of item 11125 wherein the composition is sterile.
11337. The method of any one of items 11125-11336 wherein the device is adapted for delivering insulin.
11338. The method of any one of items 11125-11336 wherein the device is adapted for delivering a narcotic.
11339. The method of any one of items 11125-11336 wherein the device is adapted for delivering a chemotherapeutic agent.
11340. The method of any one of items 11125-11336 wherein the device is adapted for delivering an anti-arrhythmic drug. 11341. The method of any one of items 11125-11336 wherein the device is adapted for delivering an anti-spasmotic drug.
11342. The method of any one of items 11125-11336 wherein the device is adapted for delivering an anti-spastic agent.
11343. The method of any one of items 11125-11336 wherein the device is adapted for delivering an antibiotic.
11344. The method of any one of items 11125-11336 wherein the device is adapted for delivering a drug only when changes in the host are detected.
11345. The method of any one of items 11125-11336 wherein the device is adapted for delivering a drug as a continuous slow release.
11346. The method of any one of items 11125-11336 wherein the device is adapted for delivering a drug at prescribed dosages in a pulsatile manner.
11347. The method of any one of items 11125-11336 wherein the device is a programmable drug delivery pump.
11348. The method of any one of items 11125-11336 wherein the device is adapted for intraocularly delivering a drug.
11349. The method of any one of items 11125-11336 wherein the device is adapted for intrathecal^ delivering a drug.
11350. The method of any one of items 11125-11336 wherein the device is adapted for intraperitoneal^ delivering a drug.
11351. The method of any one of items 11125-11336 wherein the device is adapted for intra-arterially delivering a drug. 11352. The method of any one of items 11125-11336 wherein the device is adapted for intracardiac delivery of a drug.
11353. The method of any one of items 11125-11336 wherein the device is an implantable osmotic pump.
11354. The method of any one of items 11125-11336 wherein the device is an ocular drug delivery pump.
11355. The method of any one of items 11125-11336 wherein the device is metering system.
11356. The method of any one of items 11125-11336 wherein the device is a peristaltic (roller) pump.
11357. The method of any one of items 11125-11336 wherein the device is an electronically driven pump.
11358. The method of any one of items 11125-11336 wherein the device is an elastomeric pump.
11359. The method of any one of items 11125-11336 wherein the device is a spring contraction pump.
11360. The method of any one of items 11125-11336 wherein the device is a gas-driven pump.
11361. The method of any one of items 11125-11336 wherein the device is a hydraulic pump.
11362. The method of any one of items 11125-11336 wherein the device is a piston-dependent pump.
11363. The method of any one of items 11125-11336 wherein the device is a non-piston-dependent pump. 11726
11364. The method of any one of items 11125-11336 wherein the device is a dispensing chamber.
11365. The method of any one of items 11125-11336 wherein the device is an infusion pump.
11366. The method of any one of items 11125-11336 wherein the device is a passive pump.
11367. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an implantable insulin pump.
11368. The method for implanting a medical device according to item 11367 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
11369. The method for implanting a medical device according to item 11367 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
11370. The method for implanting a medical device according to item 11367 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host. 11371. The method for implanting a medical device according to item 11367 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host.
11372. The method for implanting a medical device according to item 11367 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
11373. The method for implanting a medical device according to item 11367 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
11374. The method of item 11367 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
11375. The method of item 11367 wherein the anti-fibrotic agent is an AKT inhibitor.
11376. The method of item 11367 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11377. The method of item 11367 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
11378. The method of item 11367 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist. 11379. The method of item 11367 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVPTAEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S^836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 11380. The method of item 11367 wherein the anti-fibrotic agent is an apoptosis antagonist.
11381. The method of item 11367 wherein the anti-fibrotic agent is an apoptosis activator.
11382. The method of item 11367 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
11383. The method of item 11367 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
11384. The method of item 11367 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
11385. The method of item 11367 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
11386. The method of item 11367 wherein the anti-fibrotic agent is a calcineurin inhibitor.
11387. The method of item 11367 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
11388. The method of item 11367 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
11389. The method of item 11367 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595
(Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11390. The method of item 11367 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
11391. The method of item 11367 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
11392. The method of item 11367 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
11393. The method of item 11367 wherein the anti-fibrotic agent is a CD40 antagonist.
11394. The method of item 11367 wherein the anti-fibrotic agent is a chemokine receptor agonist.
11395. The method of item 11367 wherein the anti-fibrotic agent is a chymase inhibitor.
11396. The method of item 11367 wherein the anti-fibrotic agent is a collagenase antagonist.
11397. The method of item 11367 wherein the anti-fibrotic agent is a CXCR antagonist.
11398. The method of item 11367 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
11399. The method of item 11367 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor. 11400. The method of item 11367 wherein the anti-fibrotic agent is a DHFR inhibitor.
11401. The method of item 11367 wherein the anti-fibrotic agent is a dual integrin inhibitor.
11402. The method of item 11367 wherein the anti-fibrotic agent is an elastase inhibitor.
11403. The method of item 11367 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
11404. The method of item 11367 wherein the anti-fibrotic agent is an endothelial growth factor antagonist. .
11405. The method of item 11367 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
11406. The method of item 11367 wherein the anti-fibrotic agent is an endotoxin antagonist.
11407. The method of item 11367 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
11408. The method of item 11367 wherein the anti-fibrotic agent is an estrogen receptor antagonist. 11409. The method of item 11367 wherein the anti-fibrotic agent is an FGF inhibitor.
11410. The method of item 11367 wherein the anti-fibrotic agent is a famexyl transferase inhibitor.
11411. The method of item 11367 wherein the anti-fibrotic agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11412. The method of item 11367 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
11413. The method of item 11367 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
11414. The method of item 11367 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11415. The method of item 11367 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof. 11416. The method of item 11367 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
11417. The method of item 11367 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11418. The method of item 11367 wherein the anti-fibrotic agent is an ICAM inhibitor.
11419. The method of item 11367 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11420. The method of item 11367 wherein the anti-fibrotic agent is an IL-2 inhibitor.
11421. The method of item 11367 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
11422. The method of item 11367 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
11423. The method of item 11367 wherein the anti-fibrotic agent is an integrin antagonist.
11424. The method of item 11367 wherein the anti-fibrotic agent is an interleukin antagonist.
11425. The method of item 11367 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
11426. The method of item 11367 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11427. The method of item 11367 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
11428. The method of item 11367 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
11429. The method of item 11367 wherein the anti-fibrotic agent is a JNK inhibitor.
11430. The method of item 11367 wherein the anti-fibrotic agent is a kinase inhibitor.
11431. The method of item 11367 wherein the anti-fibrotic agent is kinesin antagonist. 11432. The method of item 11367 wherein the anti-fibrotic agent is a kinesin antagonist.
11433. The method of item 11367 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11434. The method of item 11367 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
11435. The method of item 11367 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
11436. The method of item 11367 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
11437. The method of item 11367 wherein the anti-fibrotic agent is an mTOR inhibitor.
11438. The method of item 11367 wherein the anti-fibrotic agent is an mTOR kinase inhibitor. 11439. The method of item 11367 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
11440. The method of item 11367 wherein the anti-fibrotic agent is an MIF inhibitor.
11441. The method of item 11367 wherein the anti-fibrotic agent is an MMP inhibitor.
11442. The method of item 11367 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11443. The method of item 11367 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11444. The method of item 11367 wherein the anti-fibrotic agent is a nitric oxide agonist.
11445. The method of item 11367 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
11446. The method of item 11367 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11447. The method of item 11367 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
11448. The method of item 11367 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
11449. The method of item 11367 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF- 10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11450. The method of item 11367 wherein the anti-fibrotic agent is a phosphatase inhibitor.
11451. The method of item 11367 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
11452. The method of item 11367 wherein the anti-fibrotic agent is a PKC inhibitor:
11453. The method of item 11367 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
11454. The method of item 11367 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
11455. The method of item 11367 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
11456. The method of item 11367 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
11457. The method of item 11367 wherein the anti-fibrotic agent is a protein kinase B inhibitor. 11458. The method of item 11367 wherein the anti-fibrotic agent ase C stimulant.
11459. The method of item 11367 wherein the anti-fibrotic agent cleoside analogue.
11460. The method of item 11367 wherein the anti-fibrotic agent eptor P2X antagonist.
11461. The method of item 11367 wherein the anti-fibrotic agent e inhibitor.
11462. The method of item 11367 wherein the anti-fibrotic agent inhibitor of ErbB1 and ErbB2.
11463. The method of item 11367 wherein the anti-fibrotic agent side triphosphate reductase inhibitor.
11464. The method of item 11367 wherein the anti-fibrotic agent ntagonist.
11465. The method of item 11367 wherein the anti-fibrotic agent inhibitor.
1 T466. The method of item 11367 wherein the anti-fibrotic agent ibitor.
11467. The method of item 11367 wherein the anti-fibrotic agent in inhibitor.
11468. The method of item 11367 wherein the anti-fibrotic agent se inhibitor.
11469. The method of item 11367 wherein the anti-fibrotic agent e inhibitor. 11470. The method of item 11367 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
11471. The method of item 11367 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), ^acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11472. The method of item 11367 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
11473. The method of item 11367 wherein the anti-fibrotic agent is a tubulin antagonist.
11474. The method of item 11367 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248,
SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SLM 1657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 11475. The method of item 11367 wherein the anti-fibrotic agent ibitor.
11476. The method of item 11367 wherein the anti-fibrotic agent receptor agonist.
11477. The method of item 11367 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11478. The method of item 11367 wherein the anti-fibrotic agent an mTOR inhibitor).
11479. The method of item 11367 wherein the anti-fibrotic agent (a tubulin antagonist).
11480. The method of item 11367 wherein the anti-fibrotic agent ollagenase inhibitor).
11481. The method of item 11367 wherein the anti-fibrotic agent elongation factor-1 alpha inhibitor).
11482. The method of item 11367 wherein the anti-fibrotic agent (an epithilone).
11483. The method of item 11367 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11484. The method of item 11367 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
11485. The method of item 11367 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11486. The method of item 11367 wherein the anti-fibrotic agent tin (an angiogenesis inhibitor). 11487. The method of item 11367 wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
11488. The method of item 11367 wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
11489. The method of item 11367 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
11490. The method of item 11367 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
11491. The method of item 11367 wherein the anti-infective agent is an anthracycline.
11492. The method of item 11367 wherein the anti-infective agent is doxorubicin.
11493. The method of item 11367 wherein the anti-infective agent is mitoxantrone.
11494. The method of item 11367 wherein the anti-infective agent is a fluoropyrimidine.
11495. The method of item 11367 wherein the anti-infective agent is 5-fluorouracil (5-FU).
11496. The method of item 11367 wherein the anti-infective agent is a folic acid antagonist.
11497. The method of item 11367 wherein the anti-infective agent is methotrexate.
11498. The method of item 11367 wherein the anti-infective agent is a podophylotoxin. 11499. The method of item 11367 wherein the anti-infective agent is etoposide.
11500. The method of item 11367 wherein the anti-infective agent is camptothecin.
11501. The method of item 11367 wherein the anti-infective agent is a hydroxyurea.
11502. The method of item 11367 wherein the anti-infective agent is a platinum complex.
11503. The method of item 11367 wherein the anti-infective agent is cisplatin.
11504. The method of item 11367 wherein the composition comprises an anti-thrombotic agent.
11505. The method of item 11367 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
11506. The method of item 11367 wherein the polymer is formed from reactants comprising protein.
11507. The method of item 11367 wherein the polymer is formed from reactants comprising carbohydrate.
11508. The method of item 11367 wherein the polymer is formed from reactants comprising biodegradable polymer.
11509. The method of item 11367 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
11510. The method of item 11367 wherein the polymer is formed from reactants comprising collagen. 11511. The method of item 11367 wherein the polymer is formed from reactants comprising methylated collagen.
11512. The method of item 11367 wherein the polymer is formed from reactants comprising fibrinogen.
11513. The method of item 11367 wherein the polymer is formed from reactants comprising thrombin.
11514. The method of item 11367 wherein the polymer is formed from reactants comprising blood plasma.
11515. The method of item 11367 wherein the polymer is formed from reactants comprising calcium salt.
11516. The method of item 11367 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
11517. The method of item 11367 wherein the polymer is formed from reactants comprising fibrinogen analog.
11518. The method of item 11367 wherein the polymer is formed from reactants comprising albumin.
11519. The method of item 11367 wherein the polymer is formed from reactants comprising plasminogen.
11520. The method of item 11367 wherein the polymer is formed from reactants comprising von Willebrands factor.
11521. The method of item 11367 wherein the polymer is formed from reactants comprising Factor VIII.
11522. The method of item 11367 wherein the polymer is formed from reactants comprising hypoallergenic collagen. 11523. The method of item 11367 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
11524. The method of item 11367 wherein the polymer is formed from reactants comprising telopeptide collagen.
11525. The method of item 11367 wherein the polymer is formed from reactants comprising crosslinked collagen.
11526. The method of item 11367 wherein the polymer is formed from reactants comprising aprotinin.
11527. The method of item 11367 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
11528. The method of item 11367 wherein the polymer is formed from reactants comprising gelatin.
11529. The method of item 11367 wherein the polymer is formed from reactants comprising protein conjugates.
11530. The method of item 11367 wherein the polymer is formed from reactants comprising gelatin conjugates.
11531. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic polymer.
11532. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
11533. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
11534. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups. 11535. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
11536. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
11537. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
11538. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
11539. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
11540. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
11541. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
11542. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
11543. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups. 11544. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
11545. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
11546. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
11547. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
11548. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
11549. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
11550. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
11551. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
11552. The method of item 11367 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 11553. The method of item 11367 wherein the polymer is formed from reactants comprising polylysine.
11554. The method of item 11367 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
11555. The method of item 11367 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
11556. The method of item 11367 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
11557. The method of item 11367 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
11558. The method of item 11367 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11559. The method of item 11367 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11560. The method of item 11367 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
11561. The method of item 11367 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 11562. The method of item 11367 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
11563. The method of item 11367 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
11564. The method of item 11367 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11565. The method of item 11367 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11566. The method of item 11367 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
11567. The method of item 11367 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
11568. The method of item 11367 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
11569. The method of item 11367 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
11570. The method of item 11367 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 11571. The method of item 11367 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11572. The method of item 11367 wherein the polymer is formed from reactants comprising hyaluronic acid.
11573. The method of item 11367 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
11574. The method of item 11367 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
11575. The method of item 11367 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
11576. The method of item 11367 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
11577. The method of item 11367 wherein the composition comprises a colorant.
11578. The method of item 11367 wherein the composition is sterile.
11579. The method of any one of items 11367-11578 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense mediation through the catheter.
11580. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an intrathecal drug delivery pump.
11581. The method for implanting a medical device according to item 11580 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
11582. The method for implanting a medical device according to item 11580 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
11583. The method for implanting a medical device according to item 11580 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
11584. The method for implanting a medical device according to item 11580 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host. 11585. The method for implanting a medical device according to item 11580 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
11586. The method for implanting a medical device according to item 11580 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
11587. The method of item 11580 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
11588. The method of item 11580 wherein the anti-fibrotic agent is an AKT inhibitor.
11589. The method of item 11580 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11590. The method of item 11580 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
11591. The method of item 11580 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
11592. The method of item 11580 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus
Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
11593. The method of item 11580 wherein the anti-fibrotic agent is an apoptosis antagonist.
11594. The method of item 11580 wherein the anti-fibrotic agent is an apoptosis activator. 11595. The method of item 11580 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
11596. The method of item 11580 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
11597. The method of item 11580 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
11598. The method of item 11580 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
11599. The method of item 11580 wherein the anti-fibrotic agent is a calcineurin inhibitor.
11600. The method of item 11580 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
11601. The method of item 11580 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
11602. The method of item 11580 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11603. The method of item 11580 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
11604. The method of item 11580 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
11605. The method of item 11580 wherein the anti-fibrotic agent is a cathepsin L inhibitor. 11606. The method of item 11580 wherein the anti-fibrotic agent is a CD40 antagonist.
11607. The method of item 11580 wherein the anti-fibrotic agent is a chemokine receptor agonist.
11608. The method of item 11580 wherein the anti-fibrotic agent is a chymase inhibitor.
11609. The method of item 11580 wherein the anti-fibrotic agent is a collagenase antagonist.
11610. The method of item 11580 wherein the anti-fibrotic agent is a CXCR antagonist.
11611. The method of item 11580 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
11612. The method of item 11580 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
11613. The method of item 11580 wherein the anti-fibrotic agent is a DHFR inhibitor.
11614. The method of item 11580 wherein the anti-fibrotic agent is a dual integrin inhibitor. 11615. The method of item 11580 wherein the anti-fibrotic agent is an elastase inhibitor.
11616. The method of item 11580 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
11617. The method of item 11580 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
11618. The method of item 11580 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
11619. The method of item 11580 wherein the anti-fibrotic agent is an endotoxin antagonist.
11620. The method of item 11580 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
11621. The method of item 11580 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
11622. The method of item 11580 wherein the anti-fibrotic agent is an FGF inhibitor.
11623. The method of item 11580 wherein the anti-fibrotic agent Is a famexyl transferase inhibitor. 11624. The method of item 11580 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11625. The method of item 11580 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
11626. The method of item 11580 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
11627. The method of item 11580 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11628. The method of item 11580 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo~), and an analogue or derivative thereof.
11629. The method of item 11580 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
11630. The method of item 11580 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11631. The method of item 11580 wherein the anti-fibrotic agent is an ICAM inhibitor.
11632. The method of item 11580 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11633. The method of item 11580 wherein the anti-fibrotic agent is an IL-2 inhibitor.
11634. The method of item 11580 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 11635. The method of item 11580 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
11636. The method of item 11580 wherein the anti-fibrotic agent is an integrin antagonist.
11637. The method of item 11580 wherein the anti-fibrotic agent is an interleukin antagonist.
11638. The method of item 11580 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
11639. The method of item 11580 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11640. The method of item 11580 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
11641. The method of item 11580 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
11642. The method of item 11580 wherein the anti-fibrotic agent is a JNK inhibitor.
11643. The method of item 11580 wherein the anti-fibrotic agent is a kinase inhibitor.
11644. The method of item 11580 wherein the anti-fibrotic agent is kinesin antagonist.
11645. The method of item 11580 wherein the anti-fibrotic agent is a kinesin antagonist.
11646. The method of item 11580 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11647. The method of item 11580 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
11648. The method of item 11580 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
11649. The method of item 11580 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
11650. The method of item 11580 wherein the anti-fibrotic agent is an mTOR inhibitor.
11651. The method of item 11580 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
11652. The method of item 11580 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
11653. The method of item 11580 wherein the anti-fibrotic agent is an MIF inhibitor.
11654. The method of item 11580 wherein the anti-fibrotic agent is an MMP inhibitor.
11655. The method of item 11580 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11656. The method of item 11580 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11657. The method of item 11580 wherein the anti-fibrotic agent is a nitric oxide agonist.
11658. The method of item 11580 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
11659. The method of item 11580 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11660. The method of item 11580 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
11661. The method of item 11580 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
11662. The method of item 11580 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4),
ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from
CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11663. The method of item 11580 wherein the anti-fibrotic agent is a phosphatase inhibitor.
11664. The method of item 11580 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
11665. The method of item 11580 wherein the anti-fibrotic agent is a PKC inhibitor.
11666. The method of item 11580 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
11667. The method of item 11580 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
11668. The method of item 11580 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
11669. The method of item 11580 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
11670. The method of item 11580 wherein the anti-fibrotic agent is a protein kinase B inhibitor.
11671. The method of item 11580 wherein the anti-fibrotic agent is a protein kinase C stimulant.
11672. The method of item 11580 wherein the anti-fibrotic agent is a purine nucleoside analogue.
11673. The method of item 11580 wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist. 11674. The method of item 11580 wherein the anti-fibrotic agent is a Raf kinase inhibitor.
11675. The method of item 11580 wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
11676. The method of item 11580 wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
11677. The method of item 11580 wherein the anti-fibrotic agent is an SDF-1 antagonist.
11678. The method of item 11580 wherein the anti-fibrotic agent is a sheddase inhibitor.
11679. The method of item 11580 wherein the anti-fibrotic agent is an SRC inhibitor.
11680. The method of item 11580 wherein the anti-fibrotic agent is a stromelysin inhibitor.
11681. The method of item 11580 wherein the anti-fibrotic agent is an Syk kinase inhibitor.
11682. The method of item 11580 wherein the anti-fibrotic agent is a telomerase inhibitor.
11683. The method of item 11580 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS
No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 11684. The method of item 11580 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11685. The method of item 11580 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
11686. The method of item 11580 wherein the anti-fibrotic agent is a tubulin antagonist.
11687. The method of item 11580 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (imCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
11688. The method of item 11580 wherein the anti-fibrotic agent is a VEGF inhibitor.
11689. The method of item 11580 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
11690. The method of item 11580 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor). 11691. The method of item 11580 wherein the anti-fibrotic agent an mTOR inhibitor).
11692. The method of item 11580 wherein the anti-fibrotic agent (a tubulin antagonist).
11693. The method of item 11580 wherein the anti-fibrotic agent ollagenase inhibitor).
11694. The method of item 11580 wherein the anti-fibrotic agent elongation factor-1 alpha inhibitor).
11695. The method of item 11580 wherein the anti-fibrotic agent (an epithilone).
11696. The method of item 11580 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11697. The method of item 11580 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
11698. The method of item 11580 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11699. The method of item 11580 wherein the anti-fibrotic agent tin (an angiogenesis inhibitor).
11700. The method of item 11580 wherein the anti-fibrotic agent acetate (an angiogenesis inhibitor).
11701. The method of item 11580 wherein the anti-fibrotic agent (a kinesin antagonist).
11702. The method of item 11580 wherein the anti-fibrotic agent s (an mTOR inhibitor). 11703. The method of item 11580 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
11704. The method of item 11580 wherein the anti-infective agent is an anthracycline.
11705. The method of item 11580 wherein the anti-infective agent is doxorubicin.
11706. The method of item 11580 wherein the anti-infective agent is mitoxantrone.
11707. The method of item 11580 wherein the anti-infective agent is a fluoropyrimidine.
11708. The method of item 11580 wherein the anti-infective agent is 5-fluorouracil (5-FU).
11709. The method of item 11580 wherein the anti-infective agent is a folic acid antagonist.
11710. The method of item 11580 wherein the anti-infective agent is methotrexate.
11711. The method of item 11580 wherein the anti-infective agent is a podophylotoxin.
11712. The method of item 11580 wherein the anti-infective agent is etoposide.
11713. The method of item 11580 wherein the anti-infective agent is camptothecin.
11714. The method of item 11580 wherein the anti-infective agent is a hydroxyurea. 11715. The method of item 11580 wherein the anti-infective agent is a platinum complex.
11716. The method of item 11580 wherein the anti-infective agent is cisplatin.
11717. The method of item 11580 wherein the composition comprises an anti-thrombotic agent.
11718. The method of item 11580 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
11719. The method of item 11580 wherein the polymer is formed from reactants comprising protein.
11720. The method of item 11580 wherein the polymer is formed from reactants comprising carbohydrate.
11721. The method of item 11580 wherein the polymer is formed from reactants comprising biodegradable polymer.
11722. The method of item 11580 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
11723. The method of item 11580 wherein the polymer is formed from reactants comprising collagen.
11724. The method of item 11580 wherein the polymer is formed from reactants comprising methylated collagen.
11725. The method of item 11580 wherein the polymer is formed from reactants comprising fibrinogen.
11726. The method of item 11580 wherein the polymer is formed from reactants comprising thrombin. 11727. The method of item 11580 wherein the polymer is formed from reactants comprising blood plasma.
11728. The method of item 11580 wherein the polymer is formed from reactants comprising calcium salt.
11729. The method of item 11580 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
11730. The method of item 11580 wherein the polymer is formed from reactants comprising fibrinogen analog.
11731. The method of item 11580 wherein the polymer is formed from reactants comprising albumin.
11732. The method of item 11580 wherein the polymer is formed from reactants comprising plasminogen.
11733. The method of item 11580 wherein the polymer is formed from reactants comprising von Willebrands factor.
11734. The method of item 11580 wherein the polymer is formed from reactants comprising Factor VIII.
11735. The method of item 11580 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
11736. The method of item 11580 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
11737. The method of item 11580 wherein the polymer is formed from reactants comprising telopeptide collagen.
11738. The method of item 11580 wherein the polymer is formed from reactants comprising crosslinked collagen. 11739. The method of item 11580 wherein the polymer is formed from reactants comprising aprotinin.
11740. The method of item 11580 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
11741. The method bf item 11580 wherein the polymer is formed from reactants comprising gelatin.
11742. The method of item 11580 wherein the polymer is formed from reactants comprising protein conjugates.
11743. The method of item 11580 wherein the polymer is formed from reactants comprising gelatin conjugates.
11744. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic polymer.
11745. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
11746. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
11747. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
11748. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
11749. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 11750. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
11751. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
11752. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
11753. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
11754. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
11755. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
11756. The rhethod of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
11757. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
11758. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 11759. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
11760. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
11761. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
11762. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
11763. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
11764. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
11765. The method of item 11580 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
11766. The method of item 11580 wherein the polymer is formed from reactants comprising polylysine.
11767. The method of item 11580 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion. 11768. The method of item 11580 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
11769. The method of item 11580 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
11770. The method of item 11580 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
11771. The method of item 11580 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11772. The method of item 11580 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11773. The method of item 11580 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
11774. The method of item 11580 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
11775. The method of item 11580 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
11776. The method of item 11580 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups. 11777. The method of item 11580 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11778. The method of item 11580 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11779. The method of item 11580 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
11780. The method of item 11580 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
11781. The method of item 11580 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
11782. The method of item 11580 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
11783. The method of item 11580 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11784. The method of item 11580 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11785. The method of item 11580 wherein the polymer is formed from reactants comprising hyaluronic acid. 11786. The method of item 11580 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
11787. The method of item 11580 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
11788. The method of item 11580 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
11789. The method of item 11580 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
11790. The method of item 11580 wherein the composition comprises a colorant.
11791. The method of item 11580 wherein the composition is sterile.
11792. The method of any one of items 11580-11791 wherein the device is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord.
11793. The method of any one of items 11580-11791 wherein the device is adapted for delivering a drug to the brain.
11794. The method of any one of items 11580-11791 wherein the device is adapted for intrathecal delivering baclofen. 11795. The method of any one of items 11580-11791 wherein the device further comprises an intraspinal catheter.
11796. The method of any one of items 11580-11791 further comprising a second intrathecal drug delivery pump.
11797. The method of any one of items 11580-11791 wherein the device further comprises a catheter and an electrode.
11798. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an implantable drug delivery pump for chemotherapy.
11799. The method for implanting a medical device according to item 11798 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
11800. The method for implanting a medical device according to item 11798 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
11801. The method for implanting a medical device according to item 11798 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host. 11802. The method for implanting a medical device according to item 11798 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host.
11803. The method for implanting a medical device according to item 11798 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
11804. The method for implanting a medical device according to item 11798 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
11805. The method of item 11798 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
11806. The method of item 11798 wherein the anti-fibrotic agent is an AKT inhibitor.
11807. The method of item 11798 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11808. The method of item 11798 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
11809. The method of item 11798 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist. 11810. The method of item 11798 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245
(GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 11811. The method of item 11798 wherein the anti-fibrotic agent is an apoptosis antagonist.
11812. The method of item 11798 wherein the anti-fibrotic agent is an apoptosis activator.
11813. The method of item 11798 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
11814. The method of item 11798 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
11815. The method of item 11798 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
11816. The method of item 11798 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
11817. The method of item 11798 wherein the anti-fibrotic agent is a calcineurin inhibitor.
11818. The method of item 11798 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
11819. The method of item 11798 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
11820. The method of item 11798 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595
(Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11821. The method of item 11798 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
11822. The method of item 11798 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
11823. The method of item 11798 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
11824. The method of item 11798 wherein the anti-fibrotic agent is a CD40 antagonist.
11825. The method of item 11798 wherein the anti-fibrotic agent is a chemokine receptor agonist.
11826. The method of item 11798 wherein the anti-fibrotic agent is a chymase inhibitor.
11827. The method of item 11798 wherein the anti-fibrotic agent is a collagenase antagonist.
11828. The method of item 11798 wherein the anti-fibrotic agent is a CXCR antagonist.
11829. The method of item 11798 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
11830. The method of item 11798 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor. 11831. The method of item 11798 wherein the anti-fibrotic agent is a DHFR inhibitor.
11832. The method of item 11798 wherein the anti-fibrotic agent is a dual integrin inhibitor.
11833. The method of item 11798 wherein the anti-fibrotic agent is an elastase inhibitor.
11834. The method of item 11798 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
11835. The method of item 11798 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
11836. The method of item 11798 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
11837. The method of item 11798 wherein the anti-fibrotic agent is an endotoxin antagonist.
11838. The method of item 11798 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
11839. The method of item 11798 wherein the anti-fibrotic agent is an estrogen receptor antagonist. 11840. The method of item 11798 wherein the anti-fibrotic agent is an FGF inhibitor.
11841. The method of item 11798 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor.
11842. The method of item 11798 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11843. The method of item 11798 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
11844. The method of item 11798 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
11845. The method of item 11798 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11846. The method of item 11798 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1 ',4-didehydro-1 -deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof. 11847. The method of item 11798 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
11848. The method of item 11798 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx
Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11849. The method of item 11798 wherein the anti-fibrotic agent is an ICAM inhibitor.
11850. The method of item 11798 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11851. The method of item 11798 wherein the anti-fibrotic agent is an IL-2 inhibitor.
11852. The method of item 11798 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
11853. The method of item 11798 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
11854. The method of item 11798 wherein the anti-fibrotic agent is an integrin antagonist.
11855. The method of item 11798 wherein the anti-fibrotic agent is an interleukin antagonist.
11856. The method of item 11798 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
11857. The method of item 11798 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11858. The method of item 11798 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
11859. The method of item 11798 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
11860. The method of item 11798 wherein the anti-fibrotic agent is a JNK inhibitor.
11861. The method of item 11798 wherein the anti-fibrotic agent is a kinase inhibitor.
11862. The method of item 11798 wherein the anti-fibrotic agent is kinesin antagonist. 11863. The method of item 11798 wherein the anti-fibrotic agent is a kinesin antagonist.
11864. The method of item 11798 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11865. The method of item 11798 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
11866. The method of item 11798 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
11867. The method of item 11798 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
11868. The method of item 11798 wherein the anti-fibrotic agent is an mTOR inhibitor.
11869. The method of item 11798 wherein the anti-fibrotic agent is an mTOR kinase inhibitor. 11870. The method of item 11798 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
11871. The method of item 11798 wherein the anti-fibrotic agent is an MIF inhibitor.
11872. The method of item 11798 wherein the anti-fibrotic agent is an MMP inhibitor.
11873. The method of item 11798 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11874. The method of item 11798 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11875. The method of item 11798 wherein the anti-fibrotic agent is a nitric oxide agonist.
11876. The method of item 11798 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
11877. The method of item 11798 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11878. The method of item 11798 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
11879. The method of item 11798 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
11880. The method of item 11798 wherein the anti-f ibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11881. The method of item 11798 wherein the anti-fibrotic agent is a phosphatase inhibitor.
11882. The method of item 11798 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Gienmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
11883. The method of item 11798 wherein the anti-fibrotic agent is a PKC inhibitor.
11884. The method of item 11798 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
11885. The method of item 11798 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
11886. The method of item 11798 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
11887. The method of item 11798 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
11888. The method of item 11798 wherein the anti-fibrotic agent is a protein kinase B inhibitor. 11889. The method of item 11798 wherein the anti-fibrotic agent ase C stimulant.
11890. The method of item 11798 wherein the anti-fibrotic agent cleoside analogue.
11891. The method of item 11798 wherein the anti-fibrotic agent eptor P2X antagonist.
11892. The method of item 11798 wherein the anti-fibrotic agent e inhibitor.
11893. The method of item 11798 wherein the anti-fibrotic agent inhibitor of ErbB1 and ErbB2.
11894. The method of item 11798 wherein the anti-fibrotic agent side triphosphate reductase inhibitor.
11895. The method of item 11798 wherein the anti-fibrotic agent ntagonist.
11896. The method of item 11798 wherein the anti-fibrotic agent inhibitor.
11897. The method of item 11798 wherein the anti-fibrotic agent ibitor.
11898. The method of item 11798 wherein the anti-fibrotic agent in inhibitor.
11899. The method of item 11798 wherein the anti-fibrotic agent se inhibitor.
11900. The method of item 11798 wherein the anti-fibrotic agent e inhibitor. 11901. The method of item 11798 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
11902. The method of item 11798 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical
Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11903. The method of item 11798 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
11904. The method of item 11798 wherein the anti-fibrotic agent is a tubulin antagonist.
11905. The method of item 11798 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248,
SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), Iapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 11906. The method of item 11798 wherein the anti-fibrotic agent ibitor.
11907. The method of item 11798 wherein the anti-fibrotic agent receptor agonist.
11908. The method of item 11798 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11909. The method of item 11798 wherein the anti-fibrotic agent an mTOR inhibitor).
11910. The method of item 11798 wherein the anti-fibrotic agent (a tubulin antagonist).
11911. The method of item 11798 wherein the anti-fibrotic agent ollagenase inhibitor).
11912. The method of item 11798 wherein the anti-fibrotic agent ejongation factor-1 alpha inhibitor).
11913. The method of item 11798 wherein the anti-fibrotic agent (an epithilone).
11914. The method of item 11798 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11915. The method of item 11798 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
11916. The method of item 11798 wherein the anti-fibrotic agent n angiogenesis inhibitor).
11917. The method of item 11798 wherein the anti-fibrotic agent atin (an angiogenesis inhibitor). 11918. The method of item 11798 wherein the anti-f ibrotic agent is anecortave acetate (an angiogenesis inhibitor).
11919. The method of item 11798 wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
11920. The method of item 11798 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
11921. The method of item 11798 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
11922. The method of item 11798 wherein the anti-infective agent is an anthracycline.
11923. The method of item 11798 wherein the anti-infective agent is doxorubicin.
11924. The method of item 11798 wherein the anti-infective agent is mitoxantrone.
11925. The method of item 11798 wherein the anti-infective agent is a fluoropyrimidine.
11926. The method of item 11798 wherein the anti-infective agent is 5-fluorouracil (5-FU).
11927. The method of item 11798 wherein the anti-infective agent is a folic acid antagonist.
11928. The method of item 11798 wherein the anti-infective agent is methotrexate.
11929. The method of item 11798 wherein the anti-infective agent is a podophylotoxin. 11930. The method of item 11798 wherein the anti-infective agent is etoposide.
11931. The method of item 11798 wherein the anti-infective agent is camptothecin.
11932. The method of item 11798 wherein the anti-infective agent is a hydroxyurea.
11933. The method of item 11798 wherein the anti-infective agent is a platinum complex.
11934. The method of item 11798 wherein the anti-infective agent is cisplatin.
11935. The method of item 11798 wherein the composition comprises an anti-thrombotic agent.
11936. The method of item 11798 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
11937. The method of item 11798 wherein the polymer is formed from reactants comprising protein.
11938. The method of item 11798 wherein the polymer is formed from reactants comprising carbohydrate.
11939. The method of item 11798 wherein the polymer is formed from reactants comprising biodegradable polymer.
11940. The method of item 11798 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
11941. The method of item 11798 wherein the polymer is formed from reactants comprising collagen. 11942. The method of item 11798 wherein the polymer is formed from reactants comprising methylated collagen.
11943. The method of item 11798 wherein the polymer is formed from reactants comprising fibrinogen.
11944. The method of item 11798 wherein the polymer is formed from reactants comprising thrombin.
11945. The method of item 11798 wherein the polymer is formed from reactants comprising blood plasma.
11946. The method of item 11798 wherein the polymer is formed from reactants comprising calcium salt.
11947. The method of item 11798 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
11948. The method of item 11798 wherein the polymer is formed from reactants comprising fibrinogen analog.
11949. The method of item 11798 wherein the polymer is formed from reactants comprising albumin.
11950. The method of item 11798 wherein the polymer is formed from reactants comprising plasminogen.
11951. The method of item 11798 wherein the polymer is formed from reactants comprising von Willebrands factor.
11952. The method of item 11798 wherein the polymer is formed from reactants comprising Factor VIII.
11953. The method of item 11798 wherein the polymer is formed from reactants comprising hypoallergenic collagen. 11954. The method of item 11798 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
11955. The method of item 11798 wherein the polymer is formed from reactants comprising telopeptide collagen.
11956. The method of item 11798 wherein the polymer is formed from reactants comprising crosslinked collagen.
11957. The method of item 11798 wherein the polymer is formed from reactants comprising aprotinin.
11958. The method of item 11798 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
11959. The method of item 11798 wherein the polymer is formed from reactants comprising gelatin.
11960. The method of item 11798 wherein the polymer is formed from reactants comprising protein conjugates.
11961. The method of item 11798 wherein the polymer is formed from reactants comprising gelatin conjugates.
11962. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic polymer.
11963. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
11964. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
11965. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups. 11966. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
11967. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
11968. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
11969. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
11970. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
11971. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
11972. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
11973. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
11974. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups. 11975. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
11976. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
11977. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
11978. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
11979. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
11980. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
11981. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
11982. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
11983. The method of item 11798 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 11984. The method of item 11798 wherein the polymer is formed from reactants comprising polylysine.
11985. The method of item 11798 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
11986. The method of item 11798 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
11987. The method of item 11798 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
11988. The method of item 11798 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
11989. The method of item 11798 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11990. The method of item 11798 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11991. The method of item 11798 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
11992. The method of item 11798 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 11993. The method of item 11798 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
11994. The method of item 11798 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
11995. The method of item 11798 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
11996. The method of item 11798 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
11997. The method of item 11798 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
11998. The method of item 11798 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
11999. The method of item 11798 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
12000. The method of item 11798 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
12001. The method of item 11798 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 12002. The method of item 11798 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
12003. The method of item 11798 wherein the polymer is formed from reactants comprising hyaluronic acid.
12004. The method of item 11798 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
12005. The method of item 11798 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
12006. The method of item 11798 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
12007. The method of item 11798 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
12008. The method of item 11798 wherein the composition comprises a colorant.
12009. The method of item 11798 wherein the composition is sterile.
12010. The method of any one of items 11798-12009 wherein the device is adapted for delivering 2'-deoxy 5-fluorouridine. 12011. The method of any one of items 11798-12009 wherein the host has a solid tumor, and the device is adapted for infusing a chemotherapeutic agent to the solid tumor.
12012. The method of any one of items 11798-12009 wherein the host has a tumor, and the device is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
12013. The method of any one of items 11798-12009 wherein the host has a hepatic tumor, and the device is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host.
12014. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a drug delivery pump for treating heart disease.
12015. The method for implanting a medical device according to item 12014 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
12016. The method for implanting a medical device according to item 12014 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
12017. The method for implanting a medical device according to item 12014 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
12018. The method for implanting a medical device according to item 12014 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host.
12019. The method for implanting a medical device according to item 12014 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
12020. The method for implanting a medical device according to item 12014 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
12021. The method of item 12014 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
12022. The method of item 12014 wherein the anti-fibrotic agent is an AKT inhibitor.
12023. The method of item 12014 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
12024. The method of item 12014 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist. 12025. The method of item 12014 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
12026. The method of item 12014 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GiaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Arrigen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
12027. The method of item 12014 wherein the anti-fibrotic agent is an apoptosis antagonist.
12028. The method of item 12014 wherein the anti-fibrotic agent is an apoptosis activator.
12029. The method of item 12014 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
12030. The method of item 12014 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
12031. The method of item 12014 wherein the anti-fibrotic agent is a blocker of enzyme production in. Hepatitis C.
12032. The method of item 12014 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
12033. The method of item 12014 wherein the anti-fibrotic agent is a calcineurin inhibitor.
12034. The method of item 12014 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
12035. The method of item 12014 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
12036. The method of item 12014 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
12037. The method of item 12014 wherein the anti-fibrotic agent is a cathepsin B inhibitor. 12038. The method of item 12014 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
12039. The method of item 12014 wherein the anti-fibrotic agent is a cathepsin L inhibitor.
12040. The method of item 12014 wherein the anti-fibrotic agent is a CD40 antagonist.
12041. The method of item 12014 wherein the anti-fibrotic agent is a chemokine receptor agonist.
12042. The method of item 12014 wherein the anti-fibrotic agent is a chymase inhibitor.
12043. The method of item 12014 wherein the anti-fibrotic agent is a collagenase antagonist.
12044. The method of item 12014 wherein the anti-fibrotic agent is a CXCR antagonist.
12045. The method of item 12014 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 12046. The method of item 12014 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
12047. The method of item 12014 wherein the anti-fibrotic agent is a DHFR inhibitor.
12048. The method of item 12014 wherein the anti-fibrotic agent is a dual integrin inhibitor.
12049. The method of item 12014 wherein the anti-fibrotic agent is an elastase inhibitor.
12050. The method of item 12014 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
12051. The method of item 12014 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
12052. The method of item 12014 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309
(Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
12053. The method of item 12014 wherein the anti-fibrotic agent is an endotoxin antagonist.
12054. The method of item 12014 wherein the anti-fibrotic agent is an epothilone and tubulin binder. 12055. The method of item 12014 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
12056. The method of item 12014 wherein the anti-fibrotic agent is an FGF inhibitor.
12057. The method of item 12014 wherein the anti-fibrotic agent is a farnexyl transferase inhibitor.
12058. The method of item 12014 wherein the anti-fibrotic agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme),
Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
12059. The method of item 12014 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
12060. The method of item 12014 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
12061. The method of item 12014 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11 -9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
12062. The method of item 12014 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XlV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
12063. The method of item 12014 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
12064. The method of item 12014 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
12065. The method of item 12014 wherein the anti-fibrotic agent is an ICAM inhibitor.
12066. The method of item 12014 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
12067. The method of item 12014 wherein the anti-fibrotic agent is an IL-2 inhibitor.
12068. The method of item 12014 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
12069. The method of item 12014 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
12070. The method of item 12014 wherein the anti-fibrotic agent is an integrin antagonist.
12071. The method of item 12014 wherein the anti-fibrotic agent is an interleukin antagonist.
12072. The method of item 12014 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
12073. The method of item 12014 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
12074. The method of item 12014 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
12075. The method of item 12014 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
12076. The method of item 12014 wherein the anti-fibrotic agent is a JNK inhibitor.
12077. The method of item 12014 wherein the anti-fibrotic agent is a kinase inhibitor. 12078. The method of item 12014 wherein the anti-fibrotic agent is kinesin antagonist.
12079. The method of item 12014 wherein the anti-fibrotic agent is a kinesin antagonist.
12080. The method of item 12014 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
12081. The method of item 12014 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
12082. The method of item 12014 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
12083. The method of item 12014 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
12084. The method of item 12014 wherein the anti-fibrotic agent is an mTOR inhibitor. 12085. The method of item 12014 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
12086. The method of item 12014 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore
Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
12087. The method of item 12014 wherein the anti-fibrotic agent is an MIF inhibitor.
12088. The method of item 12014 wherein the anti-fibrotic agent is an MMP inhibitor.
12089. The method of item 12014 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457
(Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 12090. The method of item 12014 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
12091. The method of item 12014 wherein the anti-fibrotic agent is a nitric oxide agonist.
12092. The method of item 12014 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
12093. The method of item 12014 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZQ-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
12094. The method of item 12014 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
12095. The method of item 12014 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-
993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
12096. The method of item 12014 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735
(GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus
Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0)
(GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
12097. The method of item 12014 wherein the anti-fibrotic agent is a phosphatase inhibitor.
12098. The method of item 12014 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
12099. The method of item 12014 wherein the anti-fibrotic agent is a PKC inhibitor.
12100. The method of item 12014 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
12101. The method of item 12014 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
12102. The method of item 12014 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
12103. The method of item 12014 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor. 12104. The method of item 12014 wherein the anti-fibrotic agent ase B inhibitor.
12105. The method of item 12014 wherein the anti-fibrotic agent ase C stimulant.
12106. The method of item 12014 wherein the anti-fibrotic agent cleoside analogue.
12107. The method of item 12014 wherein the anti-fibrotic agent eptor P2X antagonist.
12108. The method of item 12014 wherein the anti-fibrotic agent e inhibitor.
12109. The method of item 12014 wherein the anti-fibrotic agent inhibitor of ErbB1 and ErbB2.
12110. The method of item 12014 wherein the anti-fibrotic agent side triphosphate reductase inhibitor.
12111. The method of item 12014 wherein the anti-fibrotic agent ntagonist.
12112. The method of item 12014 wherein the anti-fibrotic agent inhibitor.
12113. The method of item 12014 wherein the anti-fibrotic agent ibitor.
12114. The method of item 12014 wherein the anti-fibrotic agent in inhibitor.
12115. The method of item 12014 wherein the anti-fibrotic agent se inhibitor. 12116. The method of item 12014 wherein the anti-fibrotic agent is a telomerase inhibitor.
12117. The method of item 12014 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
12118. The method of item 12014 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
12119. The method of item 12014 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
12120. The method of item 12014 wherein the anti-fibrotic agent is a tubulin antagonist.
12121. The method of item 12014 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHlR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
12122. The method of item 12014 wherein the anti-fibrotic agent is a VEGF inhibitor.
12123. The method of item 12014 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
12124. The method of item 12014 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
12125. The method of item 12014 wherein the anti-fibrotic agent is AP-23573 (an mTOR inhibitor).
12126. The method of item 12014 wherein the anti-fibrotic agent is synthadotin (a tubulin antagonist).
12127. The method of item 12014 wherein the anti-fibrotic agent is S-0885 (a collagenase inhibitor).
12128. The method of item 12014 wherein the anti-fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
12129. The method of item 12014 wherein the anti-fibrotic agent is ixabepilone (an epithilone).
12130. The method of item 12014 wherein the anti-fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
12131. The method of item 12014 wherein the anti-fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
12132. The method of item 12014 wherein the anti-fibrotic agent is ABT-518 (an angiogenesis inhibitor). 12133. The method of item 12014 wherein the anti-fibrotic agent is combretastatin (an angiogenesis inhibitor).
12134. The method of item 12014 wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
12135. The method of item 12014 wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
12136. The method of item 12014 wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
12137. The method of item 12014 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
12138. The method of item 12014 wherein the anti-infective agent is an anthracycline.
12139. The method of item 12014 wherein the anti-infective agent is doxorubicin.
12140. The method of item 12014 wherein the anti-infective agent is mitoxantrone.
12141. The method of item 12014 wherein the anti-infective agent is a fluoropyrimidine.
12142. The method of item 12014 wherein the anti-infective agent is 5-fluorouracil (5-FU).
12143. The method of item 12014 wherein the anti-infective agent is a folic acid antagonist.
12144. The method of item 12014 wherein the anti-infective agent is methotrexate. 12145. The method of item 12014 wherein the anti-infective agent is a podophylotoxin.
12146. The method of item 12014 wherein the anti-infective agent is etoposide.
12147. The method of item 12014 wherein the anti-infective agent is camptothecin.
12148. The method of item 12014 wherein the anti-infective agent is a hydroxyurea.
12149. The method of item 12014 wherein the anti-infective agent is a platinum complex.
12150. The method of item 12014 wherein the anti-infective agent is cisplatin.
12151. The method of item 12014 wherein the composition comprises an anti-thrombotic agent.
12152. The method of item 12014 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
12153. The method of item 12014 wherein the polymer is formed from reactants comprising protein.
12154. The method of item 12014 wherein the polymer is formed from reactants comprising carbohydrate.
12155. The method of item 12014 wherein the polymer is formed from reactants comprising biodegradable polymer.
12156. The method of item 12014 wherein the polymer is formed from reactants comprising nonbiodegradable polymer. 12157. The method of item 12014 wherein the polymer is formed from reactants comprising collagen.
12158. The method of item 12014 wherein the polymer is formed from reactants comprising methylated collagen.
12159. The method of item 12014 wherein the polymer is formed from reactants comprising fibrinogen.
12160. The method of item 12014 wherein the polymer is formed from reactants comprising thrombin.
12161. The method of item 12014 wherein the polymer is formed from reactants comprising blood plasma.
12162. The method of item 12014 wherein the polymer is formed from reactants comprising calcium salt.
12163. The method of item 12014 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
12164. The method of item 12014 wherein the polymer is formed from reactants comprising fibrinogen analog.
12165. The method of item 12014 wherein the polymer is formed from reactants comprising albumin.
12166. The method of item 12014 wherein the polymer is formed from reactants comprising plasminogen.
12167. The method of item 12014 wherein the polymer is formed from reactants comprising von Willebrands factor.
12168. The method of item 12014 wherein the polymer is formed from reactants comprising Factor VIII. 12169. The method of item 12014 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
12170. The method of item 12014 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
12171. The method of item 12014 wherein the polymer is formed from reactants comprising telopeptide collagen.
12172. The method of item 12014 wherein the polymer is formed from reactants comprising crosslinked collagen.
12173. The method of item 12014 wherein the polymer is formed from reactants comprising aprotinin.
12174. The method of item 12014 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
12175. The method of item 12014 wherein the polymer is formed from reactants comprising gelatin.
12176. The method of item 12014 wherein the polymer is formed from reactants comprising protein conjugates.
12177. The method of item 12014 wherein the polymer is formed from reactants comprising gelatin conjugates.
12178. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic polymer.
12179. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
12180. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 12181. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
12182. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
12183. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
12184. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
12185. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
12186. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
12187. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
12188. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
12189. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 12190. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
12191. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
12192. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
12193. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
12194. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
12195. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
12196. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
12197. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
12198. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide. 12199. The method of item 12014 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
12200. The method of item 12014 wherein the polymer is formed from reactants comprising polylysine.
12201. The method of item 12014 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
12202. The method of item 12014 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
12203. The method of item 12014 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
12204. The method of item 12014 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
12205. The method of item 12014 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
12206. The method of item 12014 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
12207. The method of item 12014 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 12208. The method of item 12014 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
12209. The method of item 12014 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
12210. The method of item 12014 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
12211. The method of item 12014 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
12212. The method of item 12014 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
12213. The method of item 12014 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
12214. The method of item 12014 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
12215. The method of item 12014 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
12216. The method of item 12014 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups. 12217. The method of item 12014 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
12218. The method of item 12014 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
12219. The method of item 12014 wherein the polymer is formed from reactants comprising hyaluronic acid.
12220. The method of item 12014 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
12221. The method of item 12014 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
12222. The method of item 12014 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
12223. The method of item 12014 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
12224. The method of item 12014 wherein the composition comprises a colorant.
12225. The method of item 12014 wherein the composition is sterile. 12226. The method of any one of items 12014-12225 wherein the device is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site.
12227. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a drug delivery implant.
12228. The method for implanting a medical device according to item 12227 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
12229. The method for implanting a medical device according to item 12227 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
12230. The method for implanting a medical device according to item 12227 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
12231. The method for implanting a medical device according to item 12227 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent and a polymer, and (b) implanting the medical device into the host. 12232. The method for implanting a medical device according to item 12227 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
12233. The method for implanting a medical device according to item 12227 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti- fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
12234. The method of item 12227 wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
12235. The method of item 12227 wherein the anti-fibrotic agent is an AKT inhibitor.
12236. The method of item 12227 wherein the anti-fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
12237. The method of item 12227 wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
12238. The method of item 12227 wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
12239. The method of item 12227 wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC- 381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris
Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD- 2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios-1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV- 201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S-137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2- methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE-8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2- sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX- 1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki- 23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
12240. The method of item 12227 wherein the anti-fibrotic agent is an apoptosis antagonist.
12241. The method of item 12227 wherein the anti-fibrotic agent is an apoptosis activator. 12242. The method of item 12227 wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
12243. The method of item 12227 wherein the anti-fibrotic agent is a beta tubulin inhibitor.
12244. The method of item 12227 wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
12245. The method of item 12227 wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
12246. The method of item 12227 wherein the anti-fibrotic agent is a calcineurin inhibitor.
12247. The method of item 12227 wherein the anti-fibrotic agent is a caspase 3 inhibitor.
12248. The method of item 12227 wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
12249. The method of item 12227 wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
12250. The method of item 12227 wherein the anti-fibrotic agent is a cathepsin B inhibitor.
12251. The method of item 12227 wherein the anti-fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
12252. The method of item 12227 wherein the anti-fibrotic agent is a cathepsin L inhibitor. 12253. The method of item 12227 wherein the anti-fibrotic agent is a CD40 antagonist.
12254. The method of item 12227 wherein the anti-fibrotic agent is a chemokine receptor agonist.
12255. The method of item 12227 wherein the anti-fibrotic agent is a chymase inhibitor.
12256. The method of item 12227 wherein the anti-fibrotic agent is a collagenase antagonist.
12257. The method of item 12227 wherein the anti-fibrotic agent is a CXCR antagonist.
12258. The method of item 12227 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
12259. The method of item 12227 wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor.
12260. The method of item 12227 wherein the anti-fibrotic agent is a DHFR inhibitor.
12261. The method of item 12227 wherein the anti-fibrotic agent is a dual integrin inhibitor. 12262. The method of item 12227 wherein the anti-fibrotic agent is an elastase inhibitor.
12263. The method of item 12227 wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
12264. The method of item 12227 wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
12265. The method of item 12227 wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
12266. The method of item 12227 wherein the anti-fibrotic agent is an endotoxin antagonist.
12267. The method of item 12227 wherein the anti-fibrotic agent is an epothilone and tubulin binder.
12268. The method of item 12227 wherein the anti-fibrotic agent is an estrogen receptor antagonist.
12269. The method of item 12227 wherein the anti-fibrotic agent is an FGF inhibitor.
12270. The method of item 12227 wherein the anti-fibrotic agent is a famexyl transferase inhibitor. 12271. The method of item 12227 wherein the anti-fibrotic agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
12272. The method of item 12227 wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
12273. The method of item 12227 wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
12274. The method of item 12227 wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
12275. The method of item 12227 wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
12276. The method of item 12227 wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
12277. The method of item 12227 wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
12278. The method of item 12227 wherein the anti-fibrotic agent is an ICAM inhibitor.
12279. The method of item 12227 wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
12280. The method of item 12227 wherein the anti-fibrotic agent is an IL-2 inhibitor.
12281. The method of item 12227 wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti-inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi- Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma- Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP- 025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 12282. The method of item 12227 wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
12283. The method of item 12227 wherein the anti-fibrotic agent is an integrin antagonist.
12284. The method of item 12227 wherein the anti-fibrotic agent is an interleukin antagonist.
12285. The method of item 12227 wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
12286. The method of item 12227 wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
12287. The method of item 12227 wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
12288. The method of item 12227 wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
12289. The method of item 12227 wherein the anti-fibrotic agent is a JNK inhibitor.
12290. The method of item 12227 wherein the anti-fibrotic agent is a kinase inhibitor.
12291. The method of item 12227 wherein the anti-fibrotic agent is kinesin antagonist.
12292. The method of item 12227 wherein the anti-fibrotic agent is a kinesin antagonist.
12293. The method of item 12227 wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), FM 12 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
12294. The method of item 12227 wherein the anti-fibrotic agent is an MAP kinase inhibitor.
12295. The method of item 12227 wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
12296. The method of item 12227 wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
12297. The method of item 12227 wherein the anti-fibrotic agent is an mTOR inhibitor.
12298. The method of item 12227 wherein the anti-fibrotic agent is an mTOR kinase inhibitor.
12299. The method of item 12227 wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi- Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
12300. The method of item 12227 wherein the anti-fibrotic agent is an MIF inhibitor.
12301. The method of item 12227 wherein the anti-fibrotic agent is an MMP inhibitor.
12302. The method of item 12227 wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis),
Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
12303. The method of item 12227 wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), bortezomib, dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS US2006/011726
No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
12304. The method of item 12227 wherein the anti-fibrotic agent is a nitric oxide agonist.
12305. The method of item 12227 wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
12306. The method of item 12227 wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
12307. The method of item 12227 wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
12308. The method of item 12227 wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
12309. The method of item 12227 wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E- 3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4),
ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol- Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46- 8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from
CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
12310. The method of item 12227 wherein the anti-fibrotic agent is a phosphatase inhibitor.
12311. The method of item 12227 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR- 24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954- 27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
12312. The method of item 12227 wherein the anti-fibrotic agent is a PKC inhibitor.
12313. The method of item 12227 wherein the anti-fibrotic agent is a platelet activating factor antagonist.
12314. The method of item 12227 wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
12315. The method of item 12227 wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
12316. The method of item 12227 wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
12317. The method of item 12227 wherein the anti-fibrotic agent is a protein kinase B inhibitor.
12318. The method of item 12227 wherein the anti-fibrotic agent is a protein kinase C stimulant.
12319. The method of item 12227 wherein the anti-fibrotic agent is a purine nucleoside analogue.
12320. The method of item 12227 wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist. 12321. The method of item 12227 wherein the anti-fibrotic agent is a Raf kinase inhibitor.
12322. The method of item 12227 wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
12323. The method of item 12227 wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
12324. The method of item 12227 wherein the anti-fibrotic agent is an SDF-1 antagonist.
12325. The method of item 12227 wherein the anti-fibrotic agent is a sheddase inhibitor.
12326. The method of item 12227 wherein the anti-fibrotic agent is an SRC inhibitor.
12327. The method of item 12227 wherein the anti-fibrotic agent is a stromelysin inhibitor.
12328. The method of item 12227 wherein the anti-fibrotic agent is an Syk kinase inhibitor.
12329. The method of item 12227 wherein the anti-fibrotic agent is a telomerase inhibitor.
12330. The method of item 12227 wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS
No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG)1 TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 12331. The method of item 12227 wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX- 4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259- 65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119
(CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC- 485 (ICOS)1 infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine
Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480
(Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13- 8) (MARNAC), RPR-132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi- Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus
Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
12332. The method of item 12227 wherein the anti-fibrotic agent is a Toll receptor inhibitor.
12333. The method of item 12227 wherein the anti-fibrotic agent is a tubulin antagonist.
12334. The method of item 12227 wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG- 13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF- 1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778
(Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8)
(GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotυzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No.
380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
12335. The method of item 12227 wherein the anti-fibrotic agent is a VEGF inhibitor.
12336. The method of item 12227 wherein the anti-fibrotic agent is a vitamin D receptor agonist.
12337. The method of item 12227 wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor). 12338. The method of item 12227 wherein the anti-fibrotic agent an mTOR inhibitor).
12339. The method of item 12227 wherein the anti-fibrotic agent (a tubulin antagonist).
12340. The method of item 12227 wherein thevanti-fibrotic agent ollagenase inhibitor).
12341. The method of item 12227 wherein the anti-fibrotic agent elongation factor-1 alpha inhibitor).
12342. The method of item 12227 wherein the anti-fibrotic agent (an epithilone).
12343. The method of item 12227 wherein the anti-fibrotic agent n angiogeriesis inhibitor).
12344. The method of item 12227 wherein the anti-fibrotic agent 5 (an angiogenesis inhibitor).
12345. The method of item 12227 wherein the anti-fibrotic agent n angiogenesis inhibitor).
12346. The method of item 12227 wherein the anti-fibrotic agent tin (an angiogenesis inhibitor).
12347. The method of item 12227 wherein the anti-fibrotic agent acetate (an angiogenesis inhibitor).
12348. The method of item 12227 wherein the anti-fibrotic agent (a kinesin antagonist).
12349. The method of item 12227 wherein the anti-fibrotic agent s (an mTOR inhibitor). 12350. The method of item 12227 wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
12351. The method of item 12227 wherein the anti-infective agent is an anthracycline.
12352. The method of item 12227 wherein the anti-infective agent is doxorubicin.
12353. The method of item 12227 wherein the anti-infective agent is mitoxantrone.
12354. The method of item 12227 wherein the anti-infective agent is a fluoropyrimidine.
12355. The method of item 12227 wherein the anti-infective agent is 5-fluorouracil (5-FU).
12356. The method of item 12227 wherein the anti-infective agent is a folic acid antagonist.
12357. The method of item 12227 wherein the anti-infective agent is methotrexate.
12358. The method of item 12227 wherein the anti-infective agent is a podophylotoxin.
12359. The method of item 12227 wherein the anti-infective agent is etoposide.
12360. The method of item 12227 wherein the anti-infective agent is camptothecin.
12361. The method of item 12227 wherein the anti-infective agent is a hydroxyurea. 12362. The method of item 12227 wherein the anti-infective agent is a platinum complex.
12363. The method of item 12227 wherein the anti-infective agent is cispiatin.
12364. The method of item 12227 wherein the composition comprises an antithrombotic agent.
12365. The method of item 12227 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
12366. The method of item 12227 wherein the polymer is formed from reactants comprising protein.
12367. The method of item 12227 wherein the polymer is formed from reactants comprising carbohydrate.
12368. The method of item 12227 wherein the polymer is formed from reactants comprising biodegradable polymer.
12369. The method of item 12227 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
12370. The method of item 12227 wherein the polymer is formed from reactants comprising collagen.
12371. The method of item 12227 wherein the polymer is formed from reactants comprising methylated collagen.
12372. The method of item 12227 wherein the polymer is formed from reactants comprising fibrinogen.
12373. The method of item 12227 wherein the polymer is formed from reactants comprising thrombin. 12374. The method of item 12227 wherein the polymer is formed from reactants comprising blood plasma.
12375. The method of item 12227 wherein the polymer is formed from reactants comprising calcium salt.
12376. The method of item 12227 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
12377. The method of item 12227 wherein the polymer is formed from reactants comprising fibrinogen analog.
12378. The method of item 12227 wherein the polymer is formed from reactants comprising albumin.
12379. The method of item 12227 wherein the polymer is formed from reactants comprising plasminogen.
12380. The method of item 12227 wherein the polymer is formed from reactants comprising von Willebrands factor.
12381. The method of item 12227 wherein the polymer is formed from reactants comprising Factor VIII.
12382. The method of item 12227 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
12383. The method of item 12227 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
12384. The method of item 12227 wherein the polymer is formed from reactants comprising telopeptide collagen.
12385. The method of item 12227 wherein the polymer is formed from reactants comprising crosslinked collagen. 12386. The method of item 12227 wherein the polymer is formed from reactants comprising aprotinin.
12387. The method of item 12227 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
12388. The method of item 12227 wherein the polymer is formed from reactants comprising gelatin.
12389. The method of item 12227 wherein the polymer is formed from reactants comprising protein conjugates.
12390. The method of item 12227 wherein the polymer is formed from reactants comprising gelatin conjugates.
12391. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic polymer.
12392. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
12393. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
12394. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
12395. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
12396. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 12397. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
12398. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
12399. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
12400. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
12401. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
12402. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
12403. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
12404. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
12405. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 12406. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
12407. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
12408. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
12409. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
12410. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
12411. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
12412. The method of item 12227 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
12413. The method of item 12227 wherein the polymer is formed from reactants comprising polylysine.
12414. The method of item 12227 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion. 12415. The method of item 12227 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
12416. The method of item 12227 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
12417. The method of item 12227 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
12418. The method of item 12227 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
12419. The method of item 12227 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
12420. The method of item 12227 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
12421. The method of item 12227 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
12422. The method of item 12227 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
12423. The method of item 12227 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups. 12424. The method of item 12227 wherein the polymer is formed from reactants comprising (a), collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
12425. The method of item 12227 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
12426. The method of item 12227 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
12427. The method of item 12227 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
12428. The method of item 12227 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
12429. The method of item 12227 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
12430. The method of item 12227 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
12431. The method of item 12227 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
12432. The method of item 12227 wherein the polymer is formed from reactants comprising hyaluronic acid. 12433. The method of item 12227 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
12434. The method of item 12227 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
12435. The method of item 12227 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
12436. The method of item 12227 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
12437. The method of item 12227 wherein the composition comprises a colorant.
12438. The method of item 12227 wherein the composition is sterile.
The present invention, in various other aspects, provides the following other itemized embodiments.
1. A device comprising (a) a sensor and (b) an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 2. The device of item 1 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
3. The device of item 1 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
4. The device of item 1 wherein the agent is mevastatin (a fibrinogen antagonist).
5. The device of item 1 wherein the agent is vincamine (a microtubule inhibitor).
6. The device of item 1 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
7. The device of item 1 wherein the agent is SKF86002 (CAS
No. 72873-74-6) (a p38 MAP kinase inhibitor).
8. The device of item 1 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
9. The device of item 1 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
10. The device of item 1 wherein the agent is a tumor necrosis factor antagonist.
11. The device of item 1 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
12. The device of item 1 wherein the agent is erucylphosphocholine.
13. The device of item 1 wherein the agent is alphastatin.
14. The device of item 1 wherein the agent is etanercept.
15. The device of item 1 wherein the agent is humicade.
16. The device of item 1 wherein the agent is gefitinib.
17. The device of item 1 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline
Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
18. The device of item 1 wherein the agent is an alpha adrenergic receptor antagonist.
19. The device of item 1 wherein the agent is an anti-psychotic compound.
20. The device of item 1 wherein the agent is a CaM kinase Il inhibitor.
21. The device of item 1 wherein the agent is a G protein agonist.
22. The device of item 1 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
23. The device of item 1 wherein the agent is an anti-microbial agent.
24. The device of item 1 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof. 25. The device of item 1 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
26. The device of item 1 wherein the agent is a D2 dopamine receptor antagonist.
27. The device of item 1 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
28. The device of item 1 wherein the agent is a dopamine antagonist.
29. The device of item 1 wherein the agent is an anesthetic compound.
30. The device of item 1 wherein the agent is a clotting factor.
31. The device of item 1 wherein the agent is a lysyl hydrolase inhibitor.
32. The device of item 1 wherein the agent is a muscarinic receptor inhibitor.
33. The device of item 1 wherein the agent is a superoxide anion generator.
34. The device of item 1 wherein the agent is a steroid. 35. The device of item 1 wherein the agent is an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
36. The device of item 1 wherein the agent is a diuretic.
37. The device of item 1 wherein the agent is an anti- coagulant.
38. The device of item 1 wherein the agent is a cyclic GMP agonist.
39. The device of item 1 wherein the agent is an adenylate cyclase agonist.
40. The device of item 1 wherein the agent is an antioxidant.
41. The device of item 1 wherein the agent is a nitric oxide synthase inhibitor.
42. The device of item 1 wherein the agent is an antineoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
43. The device of item 1 wherein the agent is a DNA synthesis inhibitor. 44. The device of item 1 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
45. The device of item 1 wherein the agent is a DNA methylation inhibitor.
46. The device of item 1 wherein the agent is a NSAID agent.
47. The device of item 1 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
48. The device of item 1 wherein the agent is an MEK1/MEK 2 inhibitor.
49. The device of item 1 wherein the agent is a NO synthase inhibitor.
50. The device of item 1 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
51. The device of item 1 wherein the agent is an ACE inhibitor.
52. The device of item 1 wherein the agent is a glycosylation inhibitor.
53. The device of item 1 wherein the agent is an intracellular calcium influx inhibitor. 54. The device of item 1 wherein the agent is an anti-emetic agent.
55. The device of item 1 wherein the agent is an acetylcholinesterase inhibitor.
56. The device of item 1 wherein the agent is an ALK-5 receptor antagonist.
57. The device of item 1 wherein the agent is a RAR/RXT antagonist.
58. The device of item 1 wherein the agent is an elF-2a inhibitor.
59. The device of item 1 wherein the agent is an S-adenosyl-L- homocysteine hydrolase inhibitor.
60. The device of item 1 wherein the agent is an estrogen agonist.
61. The device of item 1 wherein the agent is a serotonin receptor inhibitor.
62. The device of item 1 wherein the agent is an antithrombotic agent.
63. The device of item 1 wherein the agent is a tryptase inhibitor.
64. The device of item 1 wherein the agent is a pesticide. 65. The device of item 1 wherein the agent is a bone mineralization promotor.
66. The device of item 1 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
67. The device of item 1 wherein the agent is an antiinflammatory compound.
68. The device of item 1 wherein the agent is a DNA methylation promotor.
69. The device of item 1 wherein the agent is an antispasmodic agent.
70. The device of item 1 wherein the agent is a protein synthesis inhibitor.
71. The device of item 1 wherein the agent is an α-glucosidase inhibitor.
72. The device of item 1 wherein the agent is a calcium channel blocker.
73. The device of item 1 wherein the agent is a pyruvate dehydrogenase activator.
74. The device of item 1 wherein the agent is a prostaglandin inhibitor. 75. The device of item 1 wherein the agent is a sodium channel inhibitor.
76. The device of item 1 wherein the agent is a serine protease inhibitor.
77. The device of item 1 wherein the agent is an intracellular calcium flux inhibitor.
78. The device of item 1 wherein the agent is a JAK2 inhibitor.
79. The device of item 1 wherein the agent is an androgen inhibitor.
80. The device of item 1 wherein the agent is an aromatase inhibitor.
81. The device of item 1 wherein the agent is an anti-viral agent.
82. The device of item 1 wherein the agent is a 5-HT inhibitor.
83. The device of item 1 wherein the agent is an FXR antagonist.
84. The device of item 1 wherein the agent is an actin polymerization and stabilization promotor.
85. The device of item 1 wherein the agent is an AXOR12 agonist. 86. The device of item 1 wherein the agent is an angiotensin Il receptor agonist.
87. The device of item 1 wherein the agent is a platelet aggregation inhibitor.
88. The device of item 1 wherein the agent is a CB1/CB2 receptor agonist.
89. The device of item 1 wherein the agent is a norepinephrine reuptake inhibitor.
90. The device of item 1 wherein the agent is a selective serotonin reuptake inhibitor.
91. The device of item 1 wherein the agent is a reducing agent.
92. The device of item 1 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
93. The device of item 1 wherein the agent is isotretinoin.
94. The device of item 1 wherein the agent is radicicol.
95. The device of item 1 wherein the agent is clobetasol propionate.
96. The device of item 1 wherein the agent is homoharringtonine. 97. The device of item 1 wherein the agent is trichostatin A.
98. The device of item 1 wherein the agent is brefeldin A.
99. The device of item 1 wherein the agent is thapsigargin.
100. The device of item 1 wherein the agent is dolastatin 15.
101. The device of item 1 wherein the agent is cerivastatin.
102. The device of item 1 wherein the agent is jasplakinolide.
103. The device of item 1 wherein the agent is herbimycin A.
104. The device of item 1 wherein the agent is pirfenidone.
105. The device of item 1 wherein the agent is vinorelbine.
106. The device of item 1 wherein the agent is 17-DMAG.
107. The device of item 1 wherein the agent is tacrolimus.
108. The device of item 1 wherein the agent is loteprednol etabonate.
109. The device of item 1 wherein the agent is juglone.
110. The device of item 1 wherein the agent is prednisolone.
111. The device of item 1 wherein the agent is puromycin. 112. The device of item 1 wherein the agent is 3-BAABE.
113. The device of item 1 wherein the agent is cladribine.
114. The device of item 1 wherein the agent is mannose-6- phosphate.
115. The device of item 1 wherein the agent is 5-azacytidine.
116. The device of item 1 wherein the agent is Ly333531 (ruboxistaurin).
117. The device of item 1 wherein the agent is simvastatin.
118. The device of item 1, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer.
119. The device of item 1, further comprising a coating, wherein the coating comprises the anti-scarring agent.
120. The device of item 1 , further comprising a coating, wherein the coating is disposed on a surface of the device.
121. The device of item 1 , further comprising a coating, wherein the coating directly contacts the device.
122. The device of item 1 , further comprising a coating, wherein the coating indirectly contacts the device.
123. The device of item 1 , further comprising a coating, wherein the coating partially covers the device. 124. The device of item 1 , further comprising a coating, wherein the coating completely covers the device.
125. The device of item 1, further comprising a coating, wherein the coating is a uniform coating.
126. The device of item 1, further comprising a coating, wherein the coating is a non-uniform coating.
127. The device of item 1 , further comprising a coating, wherein the coating is a discontinuous coating.
128. The device of item 1, further comprising a coating, wherein the coating is a patterned coating.
129. The device of item 1, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
130. The device of item 1, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
131. The device of item 1 , further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
132. The device of item 1, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
133. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 134. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
135. The device of item 1, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
136. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
137. The device of item 1, further comprising a coating, wherein the coating further comprises a polymer.
138. The device of item 1 , further comprising a first coating having a first composition and the second coating having a second composition.
139. The device of item 1 , further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
140. The device of item 1 , further comprising a polymer.
141. The device of item 1 , further comprising a polymeric carrier.
142. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer. 143. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
144. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
145. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
146. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
147. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
148. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
149. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
150. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
151. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
152. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer. 153. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
154. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
155. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
156. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
157. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
158. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
159. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
160. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
161. The device of item 1 , further comprising a lubricious coating.
162. The device of item 1 wherein the anti-scarring agent is located within pores or holes of the device. 163. The device of item 1 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
164. The device of item 1 , further comprising a second pharmaceutically active agent.
165. The device of item 1 , further comprising an antiinflammatory agent.
166. The device of item 1 , further comprising an agent that inhibits infection.
167. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
168. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
169. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
170. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
171. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
172. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. 173. The device of item 1, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
174. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
175. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is etoposide.
176. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
177. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
178. The device of item 1, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
179. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is cisplatin.
180. The device of item 1 , further comprising an anti-thrombotic agent.
181. The device of item 1 , further comprising a visualization agent.
182. The device of item 1 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 183. The device of item 1, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
184. The device of item 1, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
185. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
186. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
187. The device of item 1, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
188. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
189. The device of item 1 , further comprising an echogenic material.
190. The device of item 1 , further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
191. The device of item 1 wherein the device is sterile.
192. The device of item 1 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 193. The device of item 1 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
194. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
195. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
196. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
197. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
198. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
199. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
200. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 201. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
202. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
203. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
204. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
205. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
206. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
207. The device of item 1 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
208. The device of item 1 wherein the device comprises about
10 μg to about 10 mg of the anti-scarring agent.
209. The device of item 1 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 210. The device of item 1 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
211. The device of item 1 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
212. The device of item 1 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
213. The device of item 1 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
214. The device of item 1 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
215. The device of item 1 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
216. The device of item 1 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
217. The device of item 1 wherein a surface of the device comprises about 1000 μig to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 218. The device of item 1 wherein the agent or the composition is affixed to the sensor.
219. The device of item 1 wherein the agent or the composition is covalently attached to the sensor.
220. The device of item 1 wherein the agent or the composition is non-covalently attached to the sensor.
221. The device of item 1 further comprising a coating that absorbs the agent or the composition.
222. The device of item 1 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
223. The device of item 1 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
224. The device of item 1 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
225. The device of item 1 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
226. The device of item 1 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
227. The device of item 1 further comprising a pump that is linked to the sensor. 228. A device comprising (a) a pump and (b) an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
229. The device of item 228 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
230. The device of item 228 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
231. The device of item 228 wherein the agent is mevastatin (a fibrinogen antagonist).
232. The device of item 228 wherein the agent is vincamine (a microtubule inhibitor).
233. The device of item 228 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
234. The device of item 228 wherein the agent is SKF86002
(CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
235. The device of item 228 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
236. The device of item 228 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
237. The device of item 228 wherein the agent is a tumor necrosis factor antagonist.
238. The device of item 228 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
239. The device of item 228 wherein the agent is erucylphosphocholine.
240. The device of item 228 wherein the agent is alphastatin.
241. The device of item 228 wherein the agent is etanercept.
242. The device of item 228 wherein the agent is humicade.
243. The device of item 228 wherein the agent is gefitinib.
244. The device of item 228 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
245. The device of item 228 wherein the agent is an alpha adrenergic receptor antagonist.
246. The device of item 228 wherein the agent is an anti- psychotic compound.
247. The device of item 228 wherein the agent is a CaM kinase Il inhibitor.
248. The device of item 228 wherein the agent is a G protein agonist.
249. The device of item 228 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
250. The device of item 228 wherein the agent is an anti- microbial agent. 251. The device of item 228 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
252. The device of item 228 wherein the agent is a thromboxane
A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
253. The device of item 228 wherein the agent is a D2 dopamine receptor antagonist
254. The device of item 228 wherein the agent is a Peptidyl- Prolyl Cis/Trans lsomerase Inhibitor.
255. The device of item 228 wherein the agent is a dopamine antagonist.
256. The device of item 228 wherein the agent is an anesthetic compound.
257. The device of item 228 wherein the agent is a clotting factor.
258. The device of item 228 wherein the agent is a lysyl hydrolase inhibitor.
259. The device of item 228 wherein the agent is a muscarinic receptor inhibitor. 260. The device of item 228 wherein the agent is a superoxide anion generator.
261. The device of item 228 wherein the agent is a steroid.
262. The device of item 228 wherein the agent is an anti- proliferative agent selected from the group consisting of silibinin (CAS No.
22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
263. The device of item 228 wherein the agent is a diuretic.
264. The device of item 228 wherein the agent is an anticoagulant.
265. The device of item 228 wherein the agent is a cyclic GMP agonist.
266. The device of item 228 wherein the agent is an adenylate cyclase agonist.
267. The device of item 228 wherein the agent is an antioxidant.
268. The device of item 228 wherein the agent is a nitric oxide synthase inhibitor.
269. The device of item 228 wherein the agent is an antineoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
270. The device of item 228 wherein the agent is a DNA synthesis inhibitor.
271. The device of item 228 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
272. The device of item 228 wherein the agent is a DNA methylation inhibitor.
273. The device of item 228 wherein the agent is a NSAID agent.
274. The device of item 228 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
275. The device of item 228 wherein the agent is an MEK1/MEK 2 inhibitor.
276. The device of item 228 wherein the agent is a NO synthase inhibitor.
277. The device of item 228 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
278. The device of item 228 wherein the agent is an ACE inhibitor. 279. The device of item 228 wherein the agent is a glycosylation inhibitor.
280. The device of item 228 wherein the agent is an intracellular calcium influx inhibitor.
281. The device of item 228 wherein the agent is an anti-emetic agent.
282. The device of item 228 wherein the agent is an acetylcholinesterase inhibitor.
283. The device of item 228 wherein the agent is an ALK-5 receptor antagonist.
284. The device of item 228 wherein the agent is a RAR/RXT antagonist.
285. The device of item 228 wherein the agent is an elF-2a inhibitor.
286. The device of item 228 wherein the agent is an S- adenosyl-L-homocysteine hydrolase inhibitor.
287. The device of item 228 wherein the agent is an estrogen agonist.
288. The device of item 228 wherein the agent is a serotonin receptor inhibitor. 289. The device of item 228 wherein the agent is an antithrombotic agent.
290. The device of item 228 wherein the agent is a tryptase inhibitor.
291. The device of item 228 wherein the agent is a pesticide.
292. The device of item 228 wherein the agent is a bone mineralization promotor.
293. The device of item 228 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
294. The device of item 228 wherein the agent is an antiinflammatory compound.
295. The device of item 228 wherein the agent is a DNA methylation promotor.
296. The device of item 228 wherein the agent is an antispasmodic agent.
297. The device of item 228 wherein the agent is a protein synthesis inhibitor.
298. The device of item 228 wherein the agent is an α- glucosidase inhibitor. 299. The device of item 228 wherein the agent is a calcium channel blocker.
300. The device of item 228 wherein the agent is a pyruvate dehydrogenase activator.
301. The device of item 228 wherein the agent is a prostaglandin inhibitor.
302. The device of item 228 wherein the agent is a sodium channel inhibitor.
303. The device of item 228 wherein the agent is a serine protease inhibitor.
304. The device of item 228 wherein the agent is an intracellular calcium flux inhibitor.
305. The device of item 228 wherein the agent is a JAK2 inhibitor.
306. The device of item 228 wherein the agent is an androgen inhibitor.
307. The device of item 228 wherein the agent is an aromatase inhibitor.
308. The device of item 228 wherein the agent is an anti-viral agent. 309. The device of item 228 wherein the agent is a 5-HT inhibitor.
310. The device of item 228 wherein the agent is an FXR antagonist.
311. The device of item 228 wherein the agent is an actin polymerization and stabilization promotor.
312. The device of item 228 wherein the agent is an AXOR12 agonist.
313. The device of item 228 wherein the agent is an angiotensin Il receptor agonist.
314. The device of item 228 wherein the agent is a platelet aggregation inhibitor.
315. The device of item 228 wherein the agent is a CB1/CB2 receptor agonist.
316. The device of item 228 wherein the agent is a norepinephrine reuptake inhibitor.
317. The device of item 228 wherein the agent is a selective serotonin reuptake inhibitor.
318. The device of item 228 wherein the agent is a reducing agent. 319. The device of item 228 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
320. The device of item 228 wherein the agent is isotretinoin.
321. The device of item 228 wherein the agent is radicicol.
322. The device of item 228 wherein the agent is clobetasol propionate.
323. The device of item 228 wherein the agent is homoharringtonine.
324. The device of item 228 wherein the agent is trichostatin A.
325. The device of item 228 wherein the agent is brefeldin A.
326. The device of item 228 wherein the agent is thapsigargin.
327. The device of item 228 wherein the agent is dolastatin 15.
328. The device of item 228 wherein the agent is cerivastatin.
329. The device of item 228 wherein the agent is jasplakinolide.
330. The device of item 228 wherein the agent is herbimycin A.
331. The device of item 228 wherein the agent is pirfenidone.
332. The device of item 228 wherein the agent is vinorelbine. 333. The device of item 228 wherein the agent is 17-DMAG.
334. The device of item 228 wherein the agent is tacrolimus.
335. The device of item 228 wherein the agent is loteprednol etabonate.
336. The device of item 228 wherein the agent is juglone.
337. The device of item 228 wherein the agent is prednisolone.
338. The device of item 228 wherein the agent is puromycin.
339. The device of item 228 wherein the agent is 3-BAABE.
340. The device of item 228 wherein the agent is cladribine.
341. The device of item 228 wherein the agent is mannose-6- phosphate.
342. The device of item 228 wherein the agent is 5-azacytidine.
343. The device of item 228 wherein the agent is Ly333531 (ruboxistaurin).
344. The device of item 228 wherein the agent is simvastatin.
345. The device of item 228, further comprising a coating, wherein the coating comprises the anti-scarring agent and a polymer. 346. The device of item 228, further comprising a coating, wherein the coating comprises the anti-scarring agent.
347. The device of item 228, further comprising a coating, wherein the coating is disposed on a surface of the device.
348. The device of item 228, further comprising a coating, wherein the coating directly contacts the device.
349. The device of item 228, further comprising a coating, wherein the coating indirectly contacts the device.
350. The device of item 228, further comprising a coating, wherein the coating partially covers the device.
351. The device of item 228, further comprising a coating, wherein the coating completely covers the device.
352. The device of item 228, further comprising a coating, wherein the coating is a uniform coating.
353. The device of item 228, further comprising a coating, wherein the coating is a non-uniform coating.
354. The device of item 228, further comprising a coating, wherein the coating is a discontinuous coating.
355. The device of item 228, further comprising a coating, wherein the coating is a patterned coating. 356. The device of item 228, further comprising a coating, wherein the coating has a thickness of 100 μm or less.
357. The device of item 228, further comprising a coating, wherein the coating has a thickness of 10 μm or less.
358. The device of item 228, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
359. The device of item 228, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
360. The device of item 228, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
361. The device of item 228, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
362. The device of item 228, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
363. The device of item 228, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
364. The device of item 228, further comprising a coating, wherein the coating further comprises a polymer. 365. The device of item 228, further comprising a first coating having a first composition and the second coating having a second composition.
366. The device of item 228, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
367. The device of item 228, further comprising a polymer.
368. The device of item 228, further comprising a polymeric carrier.
369. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
370. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
371. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
372. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
373. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
374. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 375. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
376. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
377. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
378. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
379. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
380. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
381. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
382. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
383. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
384. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 385. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
386. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
387. The device of item 228, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
388. The device of item 228, further comprising a lubricious coating.
389. The device of item 228 wherein the anti-scarring agent is located within pores or holes of the device.
390. The device of item 228 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
391. The device of item 228, further comprising a second pharmaceutically active agent.
392. The device of item 228, further comprising an antiinflammatory agent.
393. The device of item 228, further comprising an agent that inhibits infection.
394. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 395. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
396. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
397. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
398. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
399. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
400. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
401. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
402. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is etoposide.
403. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
404. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 405. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
406. The device of item 228, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
407. The device of item 228, further comprising an antithrombotic agent.
408. The device of item 228, further comprising a visualization agent.
409. The device of item 228, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
410. The device of item 228, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
411. The device of item 228, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
412. The device of item 228, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
413. The device of item 228, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 414. The device of item 228, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
415. The device of item 228, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
416. The device of item 228, further comprising an echogenic material.
417. The device of item 228, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
418. The device of item 228 wherein the device is sterile.
419. The device of item 228 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
420. The device of item 228 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
421. The device of item 228 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
422. The device of item 228 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
423. The device of item 228 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue. 424. The device of item 228 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
425. The device of item 228 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
426. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
427. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
428. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
429. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
430. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
431. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
432. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
433. The device of item 228 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
434. The device of item 228 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
435. The device of item 228 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
436. The device of item 228 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
437. The device of item 228 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
438. The device of item 228 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
439. The device of item 228 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
440. The device of item 228 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 441. The device of item 228 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
442. The device of item 228 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
443. The device of item 228 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
444. The device of item 228 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
445. The device of any one of items 1-227 wherein the sensor is a blood or tissue glucose monitor.
446. The device of any one of items 1-227 wherein the sensor is an electrolyte sensor.
447. The device of any one of items 1-227 wherein the sensor is a blood constituent sensor.
448. The device of any one of items 1-227 wherein the sensor is a temperature sensor.
449. The device of any one of items 1-227 wherein the sensor is a pH sensor. 450. The device of any one of items 1-227 wherein the sensor is an optical sensor.
451. The device of any one of items 1-227 wherein the sensor is an amperometric sensor.
452. The device of any one of items 1-227 wherein the sensor is a pressure or stress sensor.
453. The device of any one of items 1-227 wherein the sensor is a biosensor.
454. The device of any one of items 1-227 wherein the sensor is a sensing transponder.
455. The device of any one of items 1-227 wherein the sensor is a strain sensor.
456. The device of any one of items 1-227 wherein the sensor is a magnetoresistive sensor.
457. The device of any one of items 1 -227 wherein the sensor is a cardiac sensor.
458. The device of any one of items 1-227 wherein the sensor is a respiratory sensor.
459. The device of any one of items 1-227 wherein the sensor is an auditory sensor. 460. The device of any one of items 1-227 wherein the sensor is a metabolite sensor.
461. The device of any one of items 1-227 wherein the sensor detects mechanical changes.
462. The device of any one of items 1-227 wherein the sensor detects physical changes.
463. The device of any one of items 1-227 wherein the sensor detects electrochemical changes.
464. The device of any one of items 1-227 wherein the sensor detects magnetic changes.
465. The device of any one of items 1-227 wherein the sensor detects acceleration changes.
466. The device of any one of items 1-227 wherein the sensor detects ionizing radiation changes.
467. The device of any one of items 1-227 wherein the sensor detects acoustic wave changes.
468. The device of any one of items 1-227 wherein the sensor detects chemical changes.
469. The device of any one of items 1-227 wherein the sensor detects drug concentration changes. 470. The device of any one of items 1-227 wherein the sensor detects hormone changes.
471. The device of any one of items 1-227 wherein the sensor detects barometric changes.
472. The device of item 445 wherein the device is deliverable to the vascular system transluminally using a catheter on a stent platform.
473. The device of item 445 wherein the device is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
474. The device of 445 wherein the device is an electrode composed of an analyte responsive enzyme.
475. The device of item 445 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
476. The device of item 445 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin.
477. The device of item 452 wherein the device monitors blood pressure.
478. The device of item 452 wherein the device monitors fluid flow. 479. The device of item 452 wherein the device monitors pressure within an aneurysm sac.
480. The device of item 452 wherein the device monitors intracranial pressure.
481. The device of item 452 wherein the device monitors mechanical pressure associated with a bone fracture.
482. The device of item 452 wherein the device monitors barometric pressure.
483. The device of item 452 wherein the device monitors eye tremors.
484. The device of item 452 wherein the device monitors the depth of a corneal implant.
485. The device of item 452 wherein the device monitors intraocular pressure.
486. The device of item 452 wherein the device is a passive sensor with an inductor-capacitor circuit.
487. The device of item 452 wherein the device is a self- powered strain sensing system.
488. The device of item 452 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage. 489. The device of item 457 wherein the device monitors cardiac output.
490. The device of item 457 wherein the device monitors ejection fraction.
491. The device of item 457 wherein the device monitors blood pressure in a heart chamber.
492. The device of item 457 wherein the device monitors ventricular wall motions.
493. The device of item 457 wherein the device monitors blood flow to a transplanted organ.
494. The device of item 457 wherein the device monitors heart rate.
495. The device of item 458 wherein the device monitors pulmonary functions.
496. The device of item 459 wherein the device is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
497. The device of item 459 wherein the device generates an electrical audio signal.
498. The device of item 459 wherein the device is a capacitive sensor that is coupled to a vibrating auditory element. 499. The device of item 459 wherein the device is an electromagnetic sensor.
500. The device of any one of items 446 or 460 wherein the device emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
501. The device of any one of items 446 or 460 wherein the device is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader.
502. The device of any one of items 446 or 460 wherein the device is a monolithic bioelectronic device for detecting at least one analyte within the host.
503. The device of any one of items 228-444 wherein the device is adapted for delivering insulin.
504. The device of any one of items 228-444 wherein the device is adapted for delivering a narcotic.
505. The device of any one of items 228-444 wherein the device is adapted for delivering a chemotherapeutic agent.
506. The device of any one of items 228-444 wherein the device is adapted for delivering an anti-arrhythmic drug.
507. The device of any one of items 228-444 wherein the device is adapted for delivering an anti-spasmotic drug. 508. The device of any one of items 228-444 wherein the device delivering an anti-spastic agent.
509. The device of any one of items 228-444 wherein the device delivering an antibiotic.
510. The device of any one of items 228-444 wherein the device delivering a drug only when changes in the host are detected.
511. The device of any one of items 228-444 wherein the device delivering a drug as a continuous slow release.
512. The device of any one of items 228-444 wherein the device delivering a drug at prescribed dosages in a pulsatile manner.
513. The device of any one of items 228-444 wherein the device able drug delivery pump.
514. The device of any one of items 228-444 wherein the device intraocularly delivering a drug.
515. The device of any one of items 228-444 wherein the device intrathecal^ delivering a drug.
516. The device of any one of items 228-444 wherein the device intraperitoneal^ delivering a drug.
517. The device of any one of items 228-444 wherein the device intra-arterially delivering a drug. 518. The device of any one of items 228-444 wherein the device intracardiac delivery of a drug.
519. The device of any one of items 228-444 wherein the device ble osmotic pump.
520. The device of any one of items 228-444 wherein the device rug delivery pump.
521. The device of any one of items 228-444 wherein the device stem.
522. The device of any one of items 228-444 wherein the device (roller) pump.
523. The device of any one of items 228-444 wherein the device ically driven pump.
524. The device of any one of items 228-444 wherein the device ric pump.
525. The device of any one of items 228-444 wherein the device ntraction pump.
526. The device of any one of items 228-444 wherein the device n pump.
527. The device of any one of items 228-444 wherein the device pump. 528. The device of any one of items 228-444 wherein the device is a piston-dependent pump.
529. The device of any one of items 228-444 wherein the device is a non-piston-dependent pump.
530. The device of any one of items 228-444 wherein the device is a dispensing chamber.
531. The device of any one of items 228-444 wherein the device is an infusion pump.
532. The device of any one of items 228-444 wherein the device is a passive pump.
533. The device of any one of items 228-444 wherein the device is an implantable insulin pump.
534. The device of item 533 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense medication through the catheter.
535. The device of any one of items 228-444 wherein the device is an intrathecal drug delivery pump.
536. The device of item 535 wherein the device is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord.
537. The device of item 535 wherein the device is adapted for delivering a drug to the brain. 538. The device of item 535 wherein the device is adapted for intrathecal delivering baclofen.
539. The device of item 535 wherein the device further comprises an intraspinal catheter.
540. The device of item 535 further comprising a second intrathecal drug delivery pump.
541. The device of item 535 wherein the device further comprises a catheter and an electrode.
542. The device of any one of items 228-444 wherein the device is an implantable drug delivery pump for chemotherapy.
543. The device of item 542 wherein the device is adapted for delivering 2'-deoxy 5-fluorouridine.
544. The device of item 542 wherein the host has a solid tumor, and the device is adapted for infusing a chemotherapeutic agent to the solid tumor.
545. The device of item 542 wherein the host has a tumor, and the device is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
546. The device of item 542 wherein the host has a hepatic tumor, and the device is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host. 547. The device of any one of items 228-444 wherein the device is a drug delivery pump for treating heart disease.
548. The device of item 547 wherein the device is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site.
549. A method for inhibiting scarring comprising placing a sensor and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
550. The method of item 549 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
551. The method of item 549 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
552. The method of item 549 wherein the agent is mevastatin (a fibrinogen antagonist).
553. The method of item 549 wherein the agent is vincamine (a microtubule inhibitor).
554. The method of item 549 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor). 555. The method of item 549 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
556. The method of item 549 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
557. The method of item 549 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
558. The method of item 549 wherein the agent is a tumor necrosis factor antagonist.
559. The method of item 549 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
560. The method of item 549 wherein the agent is erucylphosphocholine.
561. The method of item 549 wherein the agent is alphastatin.
562. The method of item 549 wherein the agent is etanercept.
563. The method of item 549 wherein the agent is humicade. 564. The method of item 549 wherein the agent is gefitinib.
565. The method of item 549 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines {e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
566. The method of item 549 wherein the agent is an alpha adrenergic receptor antagonist.
567. The method of item 549 wherein the agent is an anti- psychotic compound.
568. The method of item 549 wherein the agent is a CaM kinase Il inhibitor.
569. The method of item 549 wherein the agent is a G protein agonist. 570. The method of item 549 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
571. The method of item 549 wherein the agent is an antimicrobial agent.
572. The method of item 549 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
573. The method of item 549 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
574. The method of item 549 wherein the agent is a D2 dopamine receptor antagonist.
575. The method of item 549 wherein the agent is a Peptidyl- Prolyl Cis/Trans lsomerase Inhibitor.
576. The method of item 549 wherein the agent is a dopamine antagonist.
577. The method of item 549 wherein the agent is an anesthetic compound. 578. The method of item 549 wherein the agent is a clotting factor.
579. The method of item 549 wherein the agent is a lysyl hydrolase inhibitor.
580. The method of item 549 wherein the agent is a muscarinic receptor inhibitor.
581. The method of item 549 wherein the agent is a superoxide anion generator.
582. The method of item 549 wherein the agent is a steroid.
583. The method of item 549 wherein the agent is an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
584. The method of item 549 wherein the agent is a diuretic.
585. The method of item 549 wherein the agent is an anticoagulant.
586. The method of item 549 wherein the agent is a cyclic GMP agonist. 587. The method of item 549 wherein the agent is an adenylate cyclase agonist.
588. The method of item 549 wherein the agent is an antioxidant.
589. The method of item 549 wherein the agent is a nitric oxide synthase inhibitor.
590. The method of item 549 wherein the agent is an antineoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
591. The method of item 549 wherein the agent is a DNA synthesis inhibitor.
592. The method of item 549 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
593. The method of item 549 wherein the agent is a DNA methylation inhibitor.
594. The method of item 549 wherein the agent is a NSAID agent.
595. The method of item 549 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor. 596. The method of item 549 wherein the agent is an MEK1/MEK 2 inhibitor.
597. The method of item 549 wherein the agent is a NO synthase inhibitor.
598. The method of item 549 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
599. The method of item 549 wherein the agent is an ACE inhibitor.
600. The method of item 549 wherein the agent is a glycosylation inhibitor.
601. The method of item 549 wherein the agent is an intracellular calcium influx inhibitor.
602. The method of item 549 wherein the agent is an anti- emetic agent.
603. The method of item 549 wherein the agent is an acetylcholinesterase inhibitor.
604. The method of item 549 wherein the agent is an ALK-5 receptor antagonist.
605. The method of item 549 wherein the agent is a RAR/RXT antagonist. 606. The method of item 549 wherein the agent is an elF-2a inhibitor.
607. The method of item 549 wherein the agent is an S- adenosyl-L-homocysteine hydrolase inhibitor.
608. The method of item 549 wherein the agent is an estrogen agonist.
609. The method of item 549 wherein the agent is a serotonin receptor inhibitor.
610. The method of item 549 wherein the agent is an anti- thrombotic agent.
611. The method of item 549 wherein the agent is a tryptase inhibitor.
612. The method of item 549 wherein the agent is a pesticide.
613. The method of item 549 wherein the agent is a bone mineralization promotor.
614. The method of item 549 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
615. The method of item 549 wherein the agent is an anti- inflammatory compound. 616. The method of item 549 wherein the agent is a DNA methylation promotor.
617. The method of item 549 wherein the agent is an antispasmodic agent.
618. The method of item 549 wherein the agent is a protein synthesis inhibitor.
619. The method of item 549 wherein the agent is an α- glucosidase inhibitor.
620. The method of item 549 wherein the agent is a calcium channel blocker.
621. The method of item 549 wherein the agent is a pyruvate dehydrogenase activator.
622. The method of item 549 wherein the agent is a prostaglandin inhibitor.
623. The method of item 549 wherein the agent is a sodium channel inhibitor.
624. The method of item 549 wherein the agent is a serine protease inhibitor.
625. The method of item 549 wherein the agent is an intracellular calcium flux inhibitor. 626. The method of item 549 wherein the agent is a JAK2 inhibitor.
627. The method of item 549 wherein the agent is an androgen inhibitor.
628. The method of item 549 wherein the agent is an aromatase inhibitor.
629. The method of item 549 wherein the agent is an anti-viral agent.
630. The method of item 549 wherein the agent is a 5-HT inhibitor.
631. The method of item 549 wherein the agent is an FXR antagonist.
632. The method of item 549 wherein the agent is an actin polymerization and stabilization promotor.
633. The method of item 549 wherein the agent is an AXOR12 agonist.
634. The method of item 549 wherein the agent is an angiotensin Il receptor agonist.
635. The method of item 549 wherein the agent is a platelet aggregation inhibitor. 636. The method of item 549 wherein the agent is a CB1/CB2 receptor agonist.
637. The method of item 549 wherein the agent is a norepinephrine reuptake inhibitor.
638. The method of item 549 wherein the agent is a selective serotonin reuptake inhibitor.
639. The method of item 549 wherein the agent is a reducing agent.
640. The method of item 549 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
641. The method of item 549 wherein the agent is isotretinoin.
642. The method of item 549 wherein the agent is radicicol.
643. The method of item 549 wherein the agent is clobetasol propionate.
644. The method of item 549 wherein the agent is homoharringtonine.
645. The method of item 549 wherein the agent is trichostatin A.
646. The method of item 549 wherein the agent is brefeldin A.
647. The method of item 549 wherein the agent is thapsigargin. 648. The method of item 549 wherein the agent is dolastatin 15.
649. The method of item 549 wherein the agent is cerivastatin.
650. The method of item 549 wherein the agent is jasplakinolide.
651. The method of item 549 wherein the agent is herbimycin A.
652. The method of item 549 wherein the agent is pirfenidone.
653. The method of item 549 wherein the agent is vinorelbine.
654. The method of item 549 wherein the agent is 17-DMAG.
655. The method of item 549 wherein the agent is tacrolimus.
656. The method of item 549 wherein the agent is loteprednol etabonate.
657. The method of item 549 wherein the agent is juglone.
658. The method of item 549 wherein the agent is prednisolone.
659. The method of item 549 wherein the agent is puromycin.
660. The method of item 549 wherein the agent is 3-BAABE.
661. The method of item 549 wherein the agent is cladribine. 662. The method of item 549 wherein the agent is mannose-6- phosphate.
663. The method of item 549 wherein the agent is 5-azacytidine.
664. The method of item 549 wherein the agent is Ly333531 (ruboxistaurin).
665. The method of item 549 wherein the agent is simvastatin.
666. The method of item 549, wherein the composition comprises a polymer.
667. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
668. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
669. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
670. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
671. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer. 672. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
673. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
674. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
675. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
676. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
677. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
678. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
679. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
680. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer. 681. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
682. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
683. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
684. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
685. The method of item 549, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
686. The method of item 549, wherein the composition further comprises a second pharmaceutically active agent.
687. The method of item 549, wherein the composition further comprises an anti-inflammatory agent.
688. The method of item 549, wherein the composition further comprises an agent that inhibits infection.
689. The method of item 549, wherein the composition further comprises an anthracycline. 690. The method of item 549, wherein the composition further comprises doxorubicin.
691. The method of item 549 wherein the composition further comprises mitoxantrone.
692. The method of item 549 wherein the composition further comprises a fluoropyrimidine.
693. The method of item 549, wherein the composition further comprises 5-fluorouracil (5-FU).
694. The method of item 549, wherein the composition further comprises a folic acid antagonist.
695. The method of item 549, wherein the composition further comprises methotrexate.
696. The method of item 549, wherein the composition further comprises a podophylotoxin.
697. The method of item 549, wherein the composition further comprises etoposide.
698. The method of item 549, wherein the composition further comprises camptothecin.
699. The method of item 549, wherein the composition further comprises a hydroxyurea. 700. The method of item 549, wherein the composition further comprises a platinum complex.
701. The method of item 549, wherein the composition further comprises cisplatin.
702. The method of item 549 wherein the composition further comprises an anti-thrombotic agent.
703. The method of item 549, wherein the composition further comprises a visualization agent.
704. The method of item 549, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
705. The method of item 549, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
706. The method of item 549, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
707. The method of item 549, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate. 708. The method of item 549, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
709. The method of item 549, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
710. The method of item 549, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
711. The method of item 549 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
712. The method of item 549 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
713. The method of item 549 wherein the composition further comprises an inflammatory cytokine.
714. The method of item 549 wherein the composition further comprises an agent that stimulates cell proliferation.
715. The method of item 549 wherein the composition further comprises a polymeric carrier. 716. The method of item 549 wherein the composition is in the form of a gel, paste, or spray.
717. The method of item 549 wherein the sensor is partially constructed with the agent or the composition.
718. The method of item 549 wherein the sensor is impregnated with the agent or the composition.
719. The method of item 549, wherein the agent or the composition forms a coating, and the coating directly contacts the sensor.
720. The method of item 549, wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor.
721. The method of item 549 wherein the agent or the composition forms a coating, and the coating partially covers the sensor.
722. The method of item 549, wherein the agent or the composition forms a coating, and the coating completely covers the sensor.
723. The method of item 549 wherein the agent or the composition is located within pores or holes of the sensor.
724. The method of item 549 wherein the agent or the composition is located within a channel, lumen, or divet of the sensor.
725. The method of item 549 wherein the sensor further comprises an echogenic material. 726. The method of item 549 wherein the sensor further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
727. The method of item 549 wherein the sensor is sterile.
728. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor.
729. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is connective tissue.
730. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is muscle tissue.
731. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is nerve tissue.
732. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the sensor after deployment of the sensor, wherein the tissue is epithelium tissue.
733. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from the time of deployment of the sensor to about 1 year. 734. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 month to 6 months.
735. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor over a period ranging from about 1 - 90 days.
736. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a constant rate.
737. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at an increasing rate.
738. The method of item 549 wherein the agent is delivered from the sensor, wherein the agent is released in effective concentrations from the sensor at a decreasing rate.
739. The method of item 549 wherein the agent is delivered from the sensor, wherein the sensor comprises about 0.01 μg to about 10 μg of the agent.
740. The method of item 549 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 μg to about 10 mg of the agent.
741. The method of item 549 wherein the agent is delivered from the sensor, wherein the sensor comprises about 10 mg to about 250 mg of the agent. 742. The method of item 549 wherein the agent is delivered from the sensor, wherein the sensor comprises about 250 mg to about 1000 mg of the agent.
743. The method of item 549 wherein the agent is delivered from the sensor, wherein the sensor comprises about 1000 mg to about 2500 mg of the agent.
744. The method of item 549 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises less than 0.01 μg of the agent per mm2 of sensor surface to which the agent is applied.
745. The method of item 549 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 0.01 μg to about 1 μg of the agent per mm2 of sensor surface to which the agent is applied.
746. The method of item 549 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1 μg to about 10 μg of the agent per mm2 of sensor surface to which the agent is applied.
747. The method of item 549 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 10 μg to about 250 μg of the agent per mm2 of sensor surface to which the agent is applied.
748. The method of item 549 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 250 μg to about 1000 μg of the agent per mm2 of sensor surface to which the agent is applied. 749. The method of item 549 wherein the agent is delivered from the sensor, wherein a surface of the sensor comprises about 1000 μg to about 2500 μg of the agent per mm2 of sensor surface to which the agent is applied.
750. The method of item 549, wherein the sensor further comprises a coating, and the coating is a uniform coating.
751. The method of item 549, wherein the sensor further comprises a coating, and the coating is a non-uniform coating.
752. The method of item 549, wherein the sensor further comprises a coating, and the coating is a discontinuous coating.
753. The method of item 549, wherein the sensor further comprises a coating, and the coating is a patterned coating.
754. The method of item 549, wherein the sensor further comprises a coating, and the coating has a thickness of 100 μm or less.
755. The method of item 549, wherein the sensor further comprises a coating, and the coating has a thickness of 10 μm or less.
756. The method of item 549, wherein the sensor further comprises a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
757. The method of item 549, wherein the sensor further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year. 758. The method of item 549, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001 % to about 1% by weight.
759. The method of item 549, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
760. The method of item 549, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
761. The method of item 549, wherein the sensor further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
762. The method of item ;549, wherein the sensor further comprises a coating, and the coating comprises a polymer.
763. The method of item 549, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition.
764. The method of item 549, wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
765. The method of item 549 wherein the agent or the composition is affixed to the sensor. 766. The method of item 549 wherein the agent or the composition is covalently attached to the sensor.
767. The method of item 549 wherein the agent or the composition is non-covalently attached to the sensor.
768. The method of item 549 wherein the sensor comprises a coating that absorbs the agent or the composition.
769. The method of item 549 wherein the sensor is interweaved with a thread composed of, or coated with, the agent or the composition.
770. The method of item 549 wherein a portion of the sensor is covered with a sleeve that contains the agent or the composition.
771. The method of item 549 wherein the sensor is completely covered with a sleeve that contains the agent or the composition.
772. The method of item 549 wherein a portion of the sensor is covered with a mesh that contains the agent or the composition.
773. The method of item 549 wherein the sensor is completely covered with a mesh that contains the agent or the composition.
774. The method of item 549 wherein the sensor is linked to a pump.
775. The method of item 549 wherein the agent or the composition is applied to the sensor surface prior to to the placing of the sensor into the host. 776. The method of item 549 wherein the agent or the composition is applied to the sensor surface during the placing of the sensor into the host.
777. The method of item 549 wherein the agent or the composition is applied to the sensor surface immediately after the placing of the sensor into the host.
778. The method of item 549 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor prior to to the placing of the sensor into the host.
779. The method of item 549 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host.
780. The method of item 549 wherein the agent or the composition is applied to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
781. The method of item 549 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed.
782. The method of item 549 wherein the agent or the composition is percutaneously injected into the tissue in the host surrounding the sensor.
783. The method of any one of items 550-782 wherein the sensor is a blood or tissue glucose monitor. 784. The method of any one of items 550-782 wherein the sensor is an electrolyte sensor.
785. The method of any one of items 550-782 wherein the sensor is a blood constituent sensor.
786. The method of any one of items 550-782 wherein the sensor is a temperature sensor.
787. The method of any one of items 550-782 wherein the sensor is a pH sensor.
788. The method of any one of items 550-782 wherein the sensor is an optical sensor.
789. The method of any one of items 550-782 wherein the sensor is an amperometric sensor.
790. The method of any one of items 550-782 wherein the sensor is a pressure or stress sensor.
791. The method of any one of items 550-782 wherein the sensor is a biosensor.
792. The method of any one of items 550-782 wherein the sensor is a sensing transponder.
793. The method of any one of items 550-782 wherein the sensor is a strain sensor. 794. The method of any one of items 550-782 wherein the sensor is a magnetoresistive sensor.
795. The method of any one of items 550-782 wherein the sensor is a cardiac sensor.
796. The method of any one of items 550-782 wherein the sensor is a respiratory sensor.
797. The method of any one of items 550-782 wherein the sensor is an auditory sensor.
798. The method of any one of items 550-782 wherein the sensor is a metabolite sensor.
799. The method of any one of items 550-782 wherein the sensor detects mechanical changes.
800. The method of any one of items 550-782 wherein the sensor detects physical changes.
801. The method of any one of items 550-782 wherein the sensor detects electrochemical changes.
802. The method of any one of items 550-782 wherein the sensor detects magnetic changes.
803. The method of any one of items 550-782 wherein the sensor detects acceleration changes. 804. The method of any one of items 550-782 wherein the sensor detects ionizing radiation changes.
805. The method of any one of items 550-782 wherein the sensor detects acoustic wave changes.
806. The method of any one of items 550-782 wherein the sensor detects chemical changes.
807. The method of any one of items 550-782 wherein the sensor detects drug concentration changes.
808. The method of any one of items 550-782 wherein the sensor detects hormone changes.
809. The method of any one of items 550-782 wherein the sensor detects barometric changes.
810. The method of item 783 wherein the device is deliverable to the vascular system transluminal^ using a catheter on a stent platform.
811. The method of item 783 wherein the device is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
812. The method of item 783 wherein the device is an electrode composed of an analyte responsive enzyme. 813. The method of item 783 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
814. The method of item 783 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin.
815. The method of item 790 wherein the device monitors blood pressure.
816. The method of item 790 wherein the device monitors fluid flow.
817. The method of item 790 wherein the device monitors pressure within an aneurysm sac.
818. The method of item 790 wherein the device monitors intracranial pressure.
819. The method of item 790 wherein the device monitors mechanical pressure associated with a bone fracture.
820. The method of item 790 wherein the device monitors barometric pressure.
821. The method of item 790 wherein the device monitors eye tremors.
822. The method of item 790 wherein the device monitors the depth of a corneal implant. 823. The method of item 790 wherein the device monitors intraocular pressure.
824. The method of item 790 wherein the device is a passive sensor with an inductor-capacitor circuit.
825. The method of item 790 wherein the device is a self- powered strain sensing system.
826. The method of item 790 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage.
827. The method of item 795 wherein the device monitors cardiac output.
828. The method of item 795 wherein the device monitors ejection fraction.
829. The method of item 795 wherein the device monitors blood pressure in a heart chamber.
830. The method of item 795 wherein the device monitors ventricular wall motions.
831. The method of item 795 wherein the device monitors blood flow to a transplanted organ.
832. The method of item 795 wherein the device monitors heart rate. 833. The method of item 796 wherein the device monitors pulmonary functions.
834. The method of item 797 wherein the device is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
835. The method of item 797 wherein the device generates an electrical audio signal.
836. The method of item 797 wherein the device is a capacitive sensor that is coupled to a vibrating auditory element.
837. The method of item 797 wherein the device is an electromagnetic sensor.
838. The method of any one of items 784 or 798 wherein the device emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
839. The method of any one of items 784 or 798 wherein the device is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader.
840. The method of any one of items 784 or 798 wherein the device is a monolithic bioelectronic device for detecting at least one analyte within the host. 841. A method for inhibiting scarring comprising placing a pump and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
842. The method of item 841 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
843. The method of item 841 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
844. The method of item 841 wherein the agent is mevastatin (a fibrinogen antagonist).
845. The method of item 841 wherein the agent is vincamine (a microtubule inhibitor).
846. The method of item 841 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
847. The method of item 841 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
848. The method of item 841 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
849. The method of item 841 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
850. The method of item 841 wherein the agent is a tumor necrosis factor antagonist.
851. The method of item 841 wherein the agent is herbimycin A
(a tyrosine kinase inhibitor).
852. The method of item 841 wherein the agent is erucylphosphocholine.
853. The method of item 841 wherein the agent is alphastatin.
854. The method of item 841 wherein the agent is etanercept.
855. The method of item 841 wherein the agent is humicade.
856. The method of item 841 wherein the agent is gefitinib.
857. The method of item 841 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co.,
Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
858. The method of item 841 wherein the agent is an alpha adrenergic receptor antagonist.
859. The method of item 841 wherein the agent is an antipsychotic compound.
860. The method of item 841 wherein the agent is a CaM kinase
Il inhibitor.
861. The method of item 841 wherein the agent is a G protein agonist.
862. The method of item 841 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
863. The method of item 841 wherein the agent is an antimicrobial agent. 864. The method of item 841 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
865. The method of item 841 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
866. The method of item 841 wherein the agent is a D2 dopamine receptor antagonist.
867. The method of item 841 wherein the agent is a Peptidyl- Prolyl Cis/Trans lsomerase Inhibitor.
868. The method of item 841 wherein the agent is a dopamine antagonist.
869. The method of item 841 wherein the agent is an anesthetic compound.
870. The method of item 841 wherein the agent is a clotting factor.
871. The method of item 841 wherein the agent is a lysyl hydrolase inhibitor.
872. The method of item 841 wherein the agent is a muscarinic receptor inhibitor. 873. The method of item 841 wherein the agent is a superoxide anion generator.
874. The method of item 841 wherein the agent is a steroid.
875. The method of item 841 wherein the agent is an anti- proliferative agent selected from the group consisting of silibinin (CAS No.
22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
876. The method of item 841 wherein the agent is a diuretic.
877. The method of item 841 wherein the agent is an anticoagulant.
878. The method of item 841 wherein the agent is a cyclic GIvIP agonist.
879. The method of item 841 wherein the agent is an adenylate cyclase agonist.
880. The method of item 841 wherein the agent is an antioxidant.
881. The method of item 841 wherein the agent is a nitric oxide synthase inhibitor. 882. The method of item 841 wherein the agent is an antineoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
883. The method of item 841 wherein the agent is a DNA synthesis inhibitor.
884. The method of item 841 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
885. The method of item 841 wherein the agent is a DNA methylation inhibitor.
886. The method of item 841 wherein the agent is a NSAID agent.
887. The method of item 841 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
888. The method of item 841 wherein the agent is an MEK1/MEK 2 inhibitor.
889. The method of item 841 wherein the agent is a NO synthase inhibitor.
890. The method of item 841 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof. 891. The method of item 841 wherein the agent is an ACE inhibitor.
892. The method of item 841 wherein the agent is a glycosylation inhibitor.
893. The method of item 841 wherein the agent is an intracellular calcium influx inhibitor.
894. The method of item 841 wherein the agent is an antiemetic agent.
895. The method of item 841 wherein the agent is an acetylcholinesterase inhibitor.
896. The method of item 841 wherein the agent is an ALK-5 receptor antagonist.
897. The method of item 841 wherein the agent is a RAR/RXT antagonist.
898. The method of item 841 wherein the agent is an elF-2a inhibitor.
899. The method of item 841 wherein the agent is an S- adenosyl-L-homocysteine hydrolase inhibitor.
900. The method of item 841 wherein the agent is an estrogen agonist. 901. The method of item 841 wherein the agent is a serotonin receptor inhibitor.
902. The method of item 841 wherein the agent is an antithrombotic agent.
903. The method of item 841 wherein the agent is a tryptase inhibitor.
904. The method of item 841 wherein the agent is a pesticide.
905. The method of item 841 wherein the agent is a bone mineralization promotor.
906. The method of item 841 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
907. The method of item 841 wherein the agent is an antiinflammatory compound.
908. The method of item 841 wherein the agent is a DNA methylation promotor.
909. The method of item 841 wherein the agent is an antispasmodic agent.
910. The method of item 841 wherein the agent is a protein synthesis inhibitor. 911. The method of item 841 wherein the agent is an α- giucosidase inhibitor.
912. The method of item 841 wherein the agent is a calcium channel blocker.
913. The method of item 841 wherein the agent is a pyruvate dehydrogenase activator.
914. The method of item 841 wherein the agent is a prostaglandin inhibitor.
915. The method of item 841 wherein the agent is a sodium channel inhibitor.
916. The method of item 841 wherein the agent is a serine protease inhibitor.
917. The method of item 841 wherein the agent is an intracellular calcium flux inhibitor.
918. The method of item 841 wherein the agent is a JAK2 inhibitor.
919. The method of item 841 wherein the agent is an androgen inhibitor.
920. The method of item 841 wherein the agent is an aromatase inhibitor. 921. The method of item 841 wherein the agent is an anti-viral agent.
922. The method of item 841 wherein the agent is a 5-HT inhibitor.
923. The method of item 841 wherein the agent is an FXR antagonist.
924. The method of item 841 wherein the agent is an actin polymerization and stabilization promotor.
925. The method of item 841 wherein the agent is an AXOR12 agonist.
926. The method of item 841 wherein the agent is an angiotensin Il receptor agonist.
927. The method of item 841 wherein the agent is a platelet aggregation inhibitor.
928. The method of item 841 wherein the agent is a CB1/CB2 receptor agonist.
929. The method of item 841 wherein the agent is a norepinephrine reuptake inhibitor.
930. The method of item 841 wherein the agent is a selective serotonin reuptake inhibitor. 931. The method of item 841 wherein the agent is a reducing agent.
932. The method of item 841 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
933. The method of item 841 wherein the agent is isotretinoin.
934. The method of item 841 wherein the agent is radicicol.
935. The method of item 841 wherein the agent is clobetasol propionate.
936. The method of item 841 wherein the agent is homoharringtonine.
937. The method of item 841 wherein the agent is trichostatin A.
938. The method of item 841 wherein the agent is brefeldin A.
939. The method of item 841 wherein the agent is thapsigargin.
940. The method of item 841 wherein the agent is dolastatin 15.
941. The method of item 841 wherein the agent is cerivastatin.
942. The method of item 841 wherein the agent is jasplakinolide.
943. The method of item 841 wherein the agent is herbimycin A. 944. The method of item 841 wherein the agent is pirfenidone.
945. The method of item 841 wherein the agent is vinorelbine.
946. The method of item 841 wherein the agent is 17-DMAG.
947. The method of item 841 wherein the agent is tacrolimus.
948. The method of item 841 wherein the agent is loteprednol etabonate.
949. The method of item 841 wherein the agent is juglone.
950. The method of item 841 wherein the agent is prednisolone.
951. The method of item 841 wherein the agent is puromycin.
952. The method of item 841 wherein the agent is 3-BAABE.
953. The method of item 841 wherein the agent is cladribine.
954. The method of item 841 wherein the agent is mannose-6- phosphate.
955. The method of item 841 wherein the agent is 5-azacytidine.
956. The method of item 841 wherein the agent is Ly333531
(ruboxistaurin).
957. The method of item 841 wherein the agent is simvastatin. 958. The method of item 841 , wherein the composition comprises a polymer.
959. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
960. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
961. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
962. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
963. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
964. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
965. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
966. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains. 967. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
968. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
969. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
970. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
971. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
972. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
973. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
974. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
975. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer. 976. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
977. The method of item 841 , wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
978. The method of item 841 , wherein the composition further comprises a second pharmaceutically active agent.
979. The method of item 841 , wherein the composition further comprises an anti-inflammatory agent.
980. The method of item 841 , wherein the composition further comprises an agent that inhibits infection.
981. The method of item 841 , wherein the composition further comprises an anthracycline.
982. The method of item 841 , wherein the composition further comprises doxorubicin.
983. The method of item 841 wherein the composition further comprises mitoxantrone.
984. The method of item 841 wherein the composition further comprises a fluoropyrimidine.
985. The method of item 841 , wherein the composition further comprises 5-fluorouracil (5-FU). 986. The method of item 841 , wherein the composition further comprises a folic acid antagonist.
987. The method of item 841 , wherein the composition further comprises methotrexate.
988. The method of item 841 , wherein the composition further comprises a podophylotoxin.
989. The method of item 841 , wherein the composition further comprises etoposide.
990. The method of item 841 , wherein the composition further comprises camptothecin.
991. The method of item 841 , wherein the composition further comprises a hydroxyurea.
992. The method of item 841 , wherein the composition further comprises a platinum complex.
993. The method of item 841 , wherein the composition further comprises cisplatin.
994. The method of item 841 wherein the composition further comprises an anti-thrombotic agent.
995. The method of item 841 , wherein the composition further comprises a visualization agent. 996. The method of item 841 , wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
997. The method of item 841 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
998. The method of item 841 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
999. The method of item 841 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
1000. The method of item 841 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
1001. The method of item 841 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
1002. The method of item 841 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant. 1003. The method of item 841 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
1004. The method of item 841 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
1005. The method of item 841 wherein the composition further comprises an inflammatory cytokine.
1006. The method of item 841 wherein the composition further comprises an agent that stimulates cell proliferation.
1007. The method of item 841 wherein the composition further comprises a polymeric carrier.
1008. The method of item 841 wherein the composition is in the form of a gel, paste, or spray.
1009. The method of any one of items 841-1008 wherein the device is adapted for delivering insulin.
1010. The method of any one of items 841-1008 wherein the device is adapted for delivering a narcotic.
1011. The method of any one of items 841-1008 wherein the device is adapted for delivering a chemotherapeutic agent. 1012. The method of any one of items 841-1008 wherein the device is adapted for delivering an anti-arrhythmic drug.
1013. The method of any one of items 841-1008 wherein the device is adapted for delivering an anti-spasmotic drug.
1014. The method of any one of items 841-1008 wherein the device is adapted for delivering an anti-spastic agent.
1015. The method of any one of items 841-1008 wherein the device is adapted for delivering an antibiotic.
1016. The method of any one of items 841-1008 wherein the device is adapted for delivering a drug only when changes in the host are detected.
1017. The method of any one of items 841-1008 wherein the device is adapted for delivering a drug as a continuous slow release.
1018. The method of any one of items 841-1008 wherein the device is adapted for delivering a drug at prescribed dosages in a pulsatile manner.
1019. The method of any one of items 841-1008 wherein the device is a programmable drug delivery pump.
1020. The method of any one of items 841-1008 wherein the device is adapted for intraocularly delivering a drug.
1021. The method of any one of items 841-1008 wherein the device is adapted for intrathecal^ delivering a drug. 1022. The method of any one of items 841-1008 wherein the device is adapted for intraperitoneally delivering a drug.
1023. The method of any one of items 841-1008 wherein the device is adapted for intra-arterially delivering a drug.
1024. The method of any one of items 841 -1008 wherein the device is adapted for intracardiac delivery of a drug.
1025. The method of any one of items 841-1008 wherein the device is an implantable osmotic pump.
1026. The method of any one of items 841-1008 wherein the device is an ocular drug delivery pump.
1027. The method of any one of items 841-1008 wherein the device is metering system.
1028. The method of any one of items 841-1008 wherein the device is a peristaltic (roller) pump.
1029. The method of any one of items 841-1008 wherein the device is an electronically driven pump.
1030. The method of any one of items 841-1008 wherein the device is an elastomeric pump.
1031. The method of any one of items 841-1008 wherein the device is a spring contraction pump. 1032. The method of any one of items 841-1008 wherein the device is a gas-driven pump.
1033. The method of any one of items 841-1008 wherein the device is a hydraulic pump.
1034. The method of any one of items 841-1008 wherein the device is a piston-dependent pump.
1035. The method of any one of items 841-1008 wherein the device is a non-piston-dependent pump.
1036. The method of any one of items 841-1008 wherein the device is a dispensing chamber.
1037. The method of any one of items 841-1008 wherein the device is an infusion pump.
1038. The method of any one of items 841-1008 wherein the device is a passive pump.
1039. The method of any one of items 841-1008 wherein the device is an implantable insulin pump.
1040. The method of item 1039 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense medication through the catheter.
1041. The method of any one of items 841-1008 wherein the device is an intrathecal drug delivery pump. 1042. The method of item 1041 wherein the device is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord.
1043. The method of item 1041 wherein the device is adapted for delivering a drug to the brain.
1044. The method of item 1041 wherein the device is adapted for intrathecal delivering baclofen.
1045. The method of item 1041 wherein the device further comprises an intraspinal catheter.
1046. The method of item 1041 further comprising a second intrathecal drug delivery pump.
1047. The method of item 1041 wherein the device further comprises a catheter and an electrode.
1048. The method of any one of items 841-1008 wherein the device is an implantable drug delivery pump for chemotherapy.
1049. The method of item 1048 wherein the device is adapted for delivering 2'-deoxy 5-fluorouridine.
1050. The method of item 1048 wherein the host has a solid tumor, and the device is adapted for infusing a chemotherapeutic agent to the solid tumor. 1051. The method of item 1048 wherein the host has a tumor, and the device is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
1052. The method of item 1048 wherein the host has a hepatic tumor, and the device is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host.
1053. The method of any one of items 841-1008 wherein the device is a drug delivery pump for treating heart disease.
1054. The method of item 1053 wherein the device is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site.
1055. A method for making a device comprising combining a sensor and an anti-scarring agent or a composition comprising anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1056. The method of item 1055 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
1057. The method of item 1055 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof. 1058. The method of item 1055 wherein the agent is mevastatin (a fibrinogen antagonist).
1059. The method of item 1055 wherein the agent is vincamine (a microtubule inhibitor).
1060. The method of item 1055 wherein the agent is emodin
(CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
1061. The method of item 1055 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
1062. The method of item 1055 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L- 165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
1063. The method of item 1055 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
1064. The method of item 1055 wherein the agent is a tumor necrosis factor antagonist.
1065. The method of item 1055 wherein the agent is herbimycin A (a tyrosine kinase inhibitor). 1066. The method of item 1055 wherein the agent is erucylphosphocholine.
1067. The method of item 1055 wherein the agent is alphastatin.
1068. The method of item 1055 wherein the agent is etanercept.
1069. The method of item 1055 wherein the agent is humicade.
1070. The method of item 1055 wherein the agent is gefitinib.
1071. The method of item 1055 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
1072. The method of item 1055 wherein the agent is an alpha adrenergic receptor antagonist. 1073. The method of item 1055 wherein the agent is an antipsychotic compound.
1074. The method of item 1055 wherein the agent is a CaM kinase Il inhibitor.
1075. The method of item 1055 wherein the agent is a G protein agonist.
1076. The method of item 1055 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
1077. The method of item 1055 wherein the agent is an antimicrobial agent.
1078. The method of item 1055 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
1079. The method of item 1055 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
1080. The method of item 1055 wherein the agent is a D2 dopamine receptor antagonist. 1081. The method of item 1055 wherein the agent is a Peptidyl- Prolyl Cis/Trans lsomerase Inhibitor.
1082. The method of item 1055 wherein the agent is a dopamine antagonist.
1083. The method of item 1055 wherein the agent is an anesthetic compound.
1084. The method of item 1055 wherein the agent is a clotting factor.
1085. The method of item 1055 wherein the agent is a lysyl hydrolase inhibitor.
1086. The method of item 1055 wherein the agent is a muscarinic receptor inhibitor.
1087. The method of item 1055 wherein the agent is a superoxide anion generator.
1088. The method of item 1055 wherein the agent is a steroid.
1089. The method of item 1055 wherein the agent is an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof. 1090. The method of item 1055 wherein the agent is a diuretic.
1091. The method of item 1055 wherein the agent is an anticoagulant.
1092. The method of item 1055 wherein the agent is a cyclic GMP agonist.
1093. The method of item 1055 wherein the agent is an adenylate cyclase agonist.
1094. The method of item 1055 wherein the agent is an antioxidant.
1095. The method of item 1055 wherein the agent is a nitric oxide synthase inhibitor.
1096. The method of item 1055 wherein the agent is an antineoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
1097. The method of item 1055 wherein the agent is a DNA synthesis inhibitor.
1098. The method of item 1055 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
1099. The method of item 1055 wherein the agent is a DNA methylation inhibitor. 1100. The method of item 1055 wherein the agent is a NSAID agent.
1101. The method of item 1055 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
1102. The method of item 1055 wherein the agent is an
MEK1/MEK 2 inhibitor.
1103. The method of item 1055 wherein the agent is a NO synthase inhibitor.
1104. The method of item 1055 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
1105. The method of item 1055 wherein the agent is an ACE inhibitor.
1106. The method of item 1055 wherein the agent is a glycosylation inhibitor.
1107. The method of item 1055 wherein the agent is an intracellular calcium influx inhibitor.
1108. The method of item 1055 wherein the agent is an antiemetic agent.
1109. The method of item 1055 wherein the agent is an acetylcholinesterase inhibitor. 1110. The method of item 1055 wherein the agent is an ALK-5 receptor antagonist.
1111. The method of item 1055 wherein the agent is a RAR/RXT antagonist.
1112. The method of item 1055 wherein the agent is an elF-2a inhibitor.
1113. The method of item 1055 wherein the agent is an S- adenosyl-L-homocysteine hydrolase inhibitor.
1114. The method of item 1055 wherein the agent is an estrogen agonist.
1115. The method of item 1055 wherein the agent is a serotonin receptor inhibitor.
1116. The method of item 1055 wherein the agent is an antithrombotic agent.
1117. The method of item 1055 wherein the agent is a tryptase inhibitor.
1118. The method of item 1055 wherein the agent is a pesticide.
1119. The method of item 1055 wherein the agent is a bone mineralization promotor. 1120. The method of item 1055 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
1121. The method of item 1055 wherein the agent is an anti- inflammatory compound.
1122. The method of item 1055 wherein the agent is a DNA methylation promotor.
1123. The method of item 1055 wherein the agent is an antispasmodic agent.
1124. The method of item 1055 wherein the agent is a protein synthesis inhibitor.
1125. The method of item 1055 wherein the agent is an α- glucosidase inhibitor.
1126. The method of item 1055 wherein the agent is a calcium channel blocker.
1127. The method of item 1055 wherein the agent is a pyruvate dehydrogenase activator.
1128. The method of item 1055 wherein the agent is a prostaglandin inhibitor.
1129. The method of item 1055 wherein the agent is a sodium channel inhibitor. 1130. The method of item 1055 wherein the agent is a serine protease inhibitor.
1131. The method of item 1055 wherein the agent is an intracellular calcium flux inhibitor.
1132. The method of item 1055 wherein the agent is a JAK2 inhibitor.
1133. The method of item 1055 wherein the agent is an androgen inhibitor.
1134. The method of item 1055 wherein the agent is an aromatase inhibitor.
1135. The method of item 1055 wherein the agent is an anti-viral agent.
1136. The method of item 1055 wherein the agent is a 5-HT inhibitor.
1137. The method of item 1055 wherein the agent is an FXR antagonist.
1138. The method of item 1055 wherein the agent is an actin polymerization and stabilization promotor.
1139. The method of item 1055 wherein the agent is an AX0R12 agonist. 1140. The method of item 1055 wherein the agent is an angiotensin Il receptor agonist.
1141. The method of item 1055 wherein the agent is a platelet aggregation inhibitor.
1142. The method of item 1055 wherein the agent is a CB1/CB2 receptor agonist.
1143. The method of item 1055 wherein the agent is a norepinephrine reuptake inhibitor.
1144. The method of item 1055 wherein the agent is a selective serotonin reuptake inhibitor.
1145. The method of item 1055 wherein the agent is a reducing agent.
1146. The method of item 1055 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
1147. The method of item 1055 wherein the agent is isotretinoin.
1148. The method of item 1055 wherein the agent is radicicol.
1149. The method of item 1055 wherein the agent is clobetasol propionate.
1150. The method of item 1055 wherein the agent is homoharringtonine. • 1151. The method of item 1055 wherein the agent is trichostatin A.
1152. The method of item 1055 wherein the agent is brefeldin A.
1153. The method of item 1055 wherein the agent is thapsigargin.
1154. The method of item 1055 wherein the agent is dolastatin 15.
1155. The method of item 1055 wherein the agent is cerivastatin.
1156. The method of item 1055 wherein the agent is jasplakinolide.
1157. The method of item 1055 wherein the agent is herbimycin A.
1158. The method of item 1055 wherein the agent is pirfenidone.
1159. The method of item 1055 wherein the agent is vinorelbine.
1160. The method of item 1055 wherein the agent is 17-DMAG.
1161. The method of item 1055 wherein the agent is tacrolimus.
1162. The method of item 1055 wherein the agent is loteprednol etabonate.
1163. The method of item 1055 wherein the agent is juglone. 1164. The method of item 1055 wherein the agent is prednisolone.
1165. The method of item 1055 wherein the agent is puromycin.
1166. The method of item 1055 wherein the agent is 3-BAABE.
1167. The method of item 1055 wherein the agent is cladribine.
1168. The method of item 1055 wherein the agent is mannose-6- phosphate.
1169. The method of item 1055 wherein the agent is 5- azacytidine.
1170. The method of item 1055 wherein the agent is Ly333531
(mboxistaurin).
1171. The method of item 1055 wherein the agent is simvastatin.
1172. The method of item 1055, wherein the composition comprises a polymer.
1173. The method of item 1055, wherein the composition comprises a polymeric carrier.
1174. The method of item 1055 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 1175. The method of item 1055 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1176. The method of item 1055 wherein the device has a coating that comprises the anti-scarring agent.
1177. The method of item 1055, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
1178. The method of item 1055, wherein the device has a coating that comprises the agent and directly contacts the sensor.
1179. The method of item 1055, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
1180. The method of item 1055, wherein the device has a coating that comprises the agent and partially covers the sensor.
1181. The method of item 1055, wherein the device has a coating that comprises the agent and completely covers the sensor.
1182. The method of item 1055, wherein the device has a uniform coating.
1183. The method of item 1055, wherein the device has a nonuniform coating.
1184. The method of item 1055, wherein the device has a discontinuous coating. 1185. The method of item 1055, wherein the device has a patterned coating.
1186. The method of item 1055, wherein the device has a coating with a thickness of 100 μm or less.
1187. The method of item 1055, wherein the device has a coating with a thickness of 10 μm or less.
1188. The method of item 1055, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
1189. The method of item 1055, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
1190. The method of item 1055, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
1191. The method of item 1055, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1192. The method of item 1055, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 1193. The method of item 1055, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1194. The method of item 1055, wherein the device has a coating, and wherein the coating further comprises a polymer.
1195. The method of item 1055, wherein the device has a first coating having a first composition and a second coating having a second composition.
1196. The method of item 1055, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
1197. The method of item 1055, wherein the composition comprises a polymer.
1198. The method of item 1055, wherein the composition comprises a polymeric carrier.
1199. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
1200. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. 1201. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
1202. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
1203. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
1204. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
1205. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
1206. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1207. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1208. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 1209. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
1210. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
1211. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
1212. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
1213. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
1214. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
1215. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
1216. The method of item 1055, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 1217. The method of item 1055 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
1218. The method of item 1055, wherein the device comprises a lubricious coating.
1219. The method of item 1055 wherein the anti-scarring agent is located within pores or holes of the device.
1220. The method of item 1055 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1221. The method of item 1055, wherein the device comprises a second pharmaceutically active agent.
1222. The method of item 1055 wherein the device comprises an anti-inflammatory agent.
1223. The method of item 1055 wherein the device comprises an agent that inhibits infection.
1224. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
1225. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
1226. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone. 1227. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
1228. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
1229. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
1230. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
1231. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
1232. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
1233. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
1234. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
1235. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
1236. The method of item 1055 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin. 1237. The method of item 1055, further comprising an antithrombotic agent.
1238. The method of item 1055 wherein the device comprises a visualization agent.
1239. The method of item 1055 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1240. The method of item 1055 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
1241. The method of item 1055 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
1242. The method of item 1055 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
1243. The method of item 1055 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1244. The method of item 1055 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 1245. The method of item 1055 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
1246. The method of item 1055 wherein the device comprises an echogenic material.
1247. The method of item 1055 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
1248. The method of item 1055 wherein the device is sterile.
1249. The method of item 1055 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1250. The method of item 1055 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
1251. The method of item 1055 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
1252. The method of item 1055 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
1253. The method of item 1055 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue. 1254. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1255. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1256. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1257. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1258. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1259. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1260. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1261. The method of item 1055 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days. 1262. The method of item 1055 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
1263. The method of item 1055 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
1264. The method of item 1055 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1265. The method of item 1055 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1266. The method of item 1055 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1267. The method of item 1055 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1268. The method of item 1055 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1269. The method of item 1055 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1270. The method of item 1055 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1271. The method of item 1055 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1272. The method of item 1055 wherein a surface of the device comprises about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1273. The method of item 1055 wherein the combining is performed by direct affixing the agent or the composition to the sensor.
1274. The method of item 1055 wherein the combining is performed by spraying the agent or the component onto the sensor.
1275. The method of item 1055 wherein the combining is performed by electrospraying the agent or the composition onto the sensor.
1276. The method of item 1055 wherein the combining is performed by dipping the sensor into a solution comprising the agent or the composition.
1277. The method of item 1055 wherein the combining is performed by covalently attaching the agent or the composition to the sensor.
1278. The method of item 1055 wherein the combining is performed by non-covalently attaching the agent or the composition to the sensor. 1279. The method of item 1055 wherein the combining is performed by coating the sensor with a substance that contains the agent or the composition.
1280. The method of item 1055 wherein the combining is performed by coating the sensor with a substance that absorbs the agent.
1281. The method of item 1055 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
1282. The method of item 1055 wherein the combining is performed by completely covering the sensor with a sleeve that contains the agent or the composition.
1283. The method of item 1055 wherein the combining is performed by covering a portion of the sensor with a sleeve that contains the agent or the composition.
1284. The method of item 1055 wherein the combining is performed by completely covering the sensor with a cover that contains the agent or the composition.
1285. The method of item 1055 wherein the combining is performed by covering a portion of the sensor with a cover that contains the agent or the composition.
1286. The method of item 1055 wherein the combining is performed by completely covering the sensor with an electrospun fabric that contains the agent or the composition. 1287. The method of item 1055 wherein the combining is performed by covering a portion of the sensor with an electrospun fabric that contains the agent or the composition.
1288. The method of item 1055 wherein the combining is performed by completely covering the sensor with a mesh that contains the agent or the composition.
1289. The method of item 1055 wherein the combining is performed by covering a portion of the sensor with a mesh that contains the agent or the composition.
1290. The method of item 1055 wherein the combining is performed by constructing a portion of the sensor with the agent or the composition.
1291. The method of item 1055 wherein the combining is performed by impregnating the sensor with the agent or the composition.
1292. The method of item 1055 wherein the combining is performed by constructing a portion of the sensor from a degradable polymer that releases the agent.
1293. The method of item 1055 wherein the combining is performed by dipping the sensor into a solution that comprise the agent and an inert solvent for the sensor.
1294. The method of item 1055 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor. 1295. The method of item 1055 wherein the combining is performed by dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
1296. The method of item 1055 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor.
1297. The method of item 1055 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
1298. The method of item 1055 wherein the combining is performed by dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
1299. The method of item 1055 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and an inert solvent for the sensor.
1300. The method of item 1055 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will swell the sensor.
1301. The method of item 1055 wherein the combining is performed by spraying the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor.
1302. The method of item 1055 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor. 1303. The method of item 1055 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor.
1304. The method of item 1055 wherein the combining is performed by spraying the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
1305. The method of any one of items 1055-1304 wherein the sensor is a blood or tissue glucose monitor.
1306. The method of any one of items 1055-1304 wherein the sensor is an electrolyte sensor.
1307. The method of any one of items 1055-1304 wherein the sensor is a blood constituent sensor.
1308. The method of any one of items 1055-1304 wherein the sensor is a temperature sensor.
1309. The method of any one of items 1055-1304 wherein the sensor is a pH sensor.
1310. The method of any one of items 1055-1304 wherein the sensor is an optical sensor.
1311. The method of any one of items 1055-1304 wherein the sensor is an amperometric sensor.
1312. The method of any one of items 1055-1304 wherein the sensor is a pressure or stress sensor. 1313. The method of any one of items 1055-1304 wherein the sensor is a biosensor.
1314. The method of any one of items 1055-1304 wherein the sensor is a sensing transponder.
1315. The method of any one of items 1055-1304 wherein the sensor is a strain sensor.
1316. The method of any one of items 1055-1304 wherein the sensor is a magnetoresistive sensor.
1317. The method of any one of items 1055-1304 wherein the sensor is a cardiac sensor.
1318. The method of any one of items 1055-1304 wherein the sensor is a respiratory sensor.
1319. The method of any one of items 1055-1304 wherein the sensor is an auditory sensor.
1320. The method of any one of items 1055-1304 wherein the sensor is a metabolite sensor.
1321. The method of any one of items 1055-1304 wherein the sensor detects mechanical changes.
1322. The method of any one of items 1055-1304 wherein the sensor detects physical changes. 1323. The method of any one of items 1055-1304 wherein the sensor detects electrochemical changes.
1324. The method of any one of items 1055-1304 wherein the sensor detects magnetic changes.
1325. The method of any one of items 1055-1304 wherein the sensor detects acceleration changes.
1326. The method of any one of items 1055-1304 wherein the sensor detects ionizing radiation changes.
1327. The method of any one of items 1055-1304 wherein the sensor detects acoustic wave changes.
1328. The method of any one of items 1055-1304 wherein the sensor detects chemical changes.
1329. The method of any one of items 1055-1304 wherein the sensor detects drug concentration changes.
1330. The method of any one of items 1055-1304 wherein the sensor detects hormone changes.
1331. The method of any one of items 1055-1304 wherein the sensor detects barometric changes.
1332. The method of item 1305 wherein the device is deliverable to the vascular system transluminal^/ using a catheter on a stent platform. 1333. The method of item 1305 wherein the device is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
1334. The method of item 1305 wherein the device is an electrode composed of an analyte responsive enzyme.
1335. The method of item 1305 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
1336. The method of item 1305 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin.
1337. The method of item 1312 wherein the device monitors blood pressure.
1338. The method of item 1312 wherein the device monitors fluid flow.
1339. The method of item 1312 wherein the device monitors pressure within an aneurysm sac.
1340. The method of item 1312 wherein the device monitors intracranial pressure.
1341. The method of item 1312 wherein the device monitors mechanical pressure associated with a bone fracture. 1342. The method of item 1312 wherein the device monitors barometric pressure.
1343. The method of item 1312 wherein the device monitors eye tremors.
1344. The method of item 1312 wherein the device monitors the depth of a corneal implant.
1345. The method of item 1312 wherein the device monitors intraocular pressure.
1346. The method of item 1312 wherein the device is a passive sensor with an inductor-capacitor circuit.
1347. The method of item 1312 wherein the device is a self- powered strain sensing system.
1348. The method of item 1312 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage.
1349. The method of item 1317 wherein the device monitors cardiac output.
1350. The method of item 1317 wherein the device monitors ejection fraction.
1351. The method of item 1317 wherein the device monitors blood pressure in a heart chamber. 1352. The method of item 1317 wherein the device monitors ventricular wall motions.
1353. The method of item 1317 wherein the device monitors blood flow to a transplanted organ.
1354. The method of item 1317 wherein the device monitors heart rate.
1355. The method of item 1318 wherein the device monitors pulmonary functions.
1356. The method of item 1319 wherein the device is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
1357. The method of item 1319 wherein the device generates an electrical audio signal.
1358. The method of item 1319 wherein the device is a capacitive sensor that is coupled to a vibrating auditory element.
1359. The method of item 1319 wherein the device is an electromagnetic sensor.
1360. The method of any one of items 1312 or 1320 wherein the device emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
1361. The method of any one of items 1312 or 1320 wherein the device is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader.
1362. The method of any one of items 1312 or 1320 wherein the device is a monolithic bioelectronic device for detecting at least one analyte within the host.
1363. A method for making a device comprising combining a pump and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1364. The method of item 1363 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
1365. The method of item 1363 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
1366. The method of item 1363 wherein the agent is mevastatin (a fibrinogen antagonist).
1367. The method of item 1363 wherein the agent is vincamine (a microtubule inhibitor). 1368. The method of item 1363 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
1369. The method of item 1363 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
1370. The method of item 1363 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
1371. The method of item 1363 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
1372. The method of item 1363 wherein the agent is a tumor necrosis factor antagonist.
1373. The method of item 1363 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
1374. The method of item 1363 wherein the agent is erucylphosphocholine.
1375. The method of item 1363 wherein the agent is alphastatin. 1376. The method of item 1363 wherein the agent is etanercept.
1377. The method of item 1363 wherein the agent is humicade.
1378. The method of item 1363 wherein the agent is gefitinib.
1379. The method of item 1363 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline
Beecham Phamaceutical Co., Wilmington, DE)1 ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
1380. The method of item 1363 wherein the agent is an alpha adrenergic receptor antagonist.
1381. The method of item 1363 wherein the agent is an antipsychotic compound.
1382. The method of item 1363 wherein the agent is a CaM kinase Il inhibitor. 1383. The method of item 1363 wherein the agent is a G protein agonist.
1384. The method of item 1363 wherein the agent is an antibiotic selected from the group consisting of apigenin (Gas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
1385. The method of item 1363 wherein the agent is an antimicrobial agent.
1386. The method of item 1363 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone
(CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
1387. The method of item 1363 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
1388. The method of item 1363 wherein the agent is a D2 dopamine receptor antagonist.
1389. The method of item 1363 wherein the agent is a Peptidyl- Prolyl Cis/Trans lsomerase Inhibitor.
1390. The method of item 1363 wherein the agent is a dopamine antagonist. 1391. The method of item 1363 wherein the agent is an anesthetic compound.
1392. The method of item 1363 wherein the agent is a clotting factor.
1393. The method of item 1363 wherein the agent is a lysyl hydrolase inhibitor.
1394. The method of item 1363 wherein the agent is a muscarinic receptor inhibitor.
1395. The method of item 1363 wherein the agent is a superoxide anion generator.
1396. The method of item 1363 wherein the agent is a steroid.
1397. The method of item 1363 wherein the agent is an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
1398. The method of item 1363 wherein the agent is a diuretic.
1399. The method of item 1363 wherein the agent is an anticoagulant. 1400. The method of item 1363 wherein the agent is a cyclic GMP agonist.
1401. The method of item 1363 wherein the agent is an adenylate cyclase agonist.
1402. The method of item 1363 wherein the agent is an antioxidant.
1403. The method of item 1363 wherein the agent is a nitric oxide synthase inhibitor.
1404. The method of item 1363 wherein the agent is an anti- neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
1405. The method of item 1363 wherein the agent is a DNA synthesis inhibitor.
1406. The method of item 1363 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
1407. The method of item 1363 wherein the agent is a DNA methylation inhibitor.
1408. The method of item 1363 wherein the agent is a NSAID agent. 1409. The method of item 1363 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
1410. The method of item 1363 wherein the agent is an MEK1/MEK 2 inhibitor.
1411. The method of item 1363 wherein the agent is a NO synthase inhibitor.
1412. The method of item 1363 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
1413. The method of item 1363 wherein the agent is an ACE inhibitor.
1414. The method of item 1363 wherein the agent is a glycosylation inhibitor.
1415. The method of item 1363 wherein the agent is an intracellular calcium influx inhibitor.
1416. The method of item 1363 wherein the agent is an antiemetic agent.
1417. The method of item 1363 wherein the agent is an acetylcholinesterase inhibitor.
1418. The method of item 1363 wherein the agent is an ALK-5 receptor antagonist. 1419. The method of item 1363 wherein the agent is a RAR/RXT antagonist.
1420. The method of item 1363 wherein the agent is an elF-2a inhibitor.
1421. The method of item 1363 wherein the agent is an S- adenosyl-L-homocysteine hydrolase inhibitor.
1422. The method of item 1363 wherein the agent is an estrogen agonist.
1423. The method of item 1363 wherein the agent is a serotonin receptor inhibitor.
1424. The method of item 1363 wherein the agent is an antithrombotic agent.
1425. The method of item 1363 wherein the agent is a tryptase inhibitor.
1426. The method of item 1363 wherein the agent is a pesticide.
1427. The method of item 1363 wherein the agent is a bone mineralization promotor.
1428. The method of item 1363 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof. 1429. The method of item 1363 wherein the agent is an antiinflammatory compound.
1430. The method of item 1363 wherein the agent is a DNA methylation promotor.
1431. The method of item 1363 wherein the agent is an antispasmodic agent.
1432. The method of item 1363 wherein the agent is a protein synthesis inhibitor.
1433. The method of item 1363 wherein the agent is an α- glucosidase inhibitor.
1434. The method of item 1363 wherein the agent is a calcium channel blocker.
1435. The method of item 1363 wherein the agent is a pyruvate dehydrogenase activator.
1436. The method of item 1363 wherein the agent is a prostaglandin inhibitor.
1437. The method of item 1363 wherein the agent is a sodium channel inhibitor.
1438. The method of item 1363 wherein the agent is a serine protease inhibitor. 1439. The method of item 1363 wherein the agent is an intracellular calcium flux inhibitor.
1440. The method of item 1363 wherein the agent is a JAK2 inhibitor.
1441. The method of item 1363 wherein the agent is an androgen inhibitor.
1442. The method of item 1363 wherein the agent is an aromatase inhibitor.
1443. The method of item 1363 wherein the agent is an anti-viral agent.
1444. The method of item 1363 wherein the agent is a 5-HT inhibitor.
1445. The method of item 1363 wherein the agent is an FXR antagonist.
1446. The method of item 1363 wherein the agent is an actin polymerization and stabilization promotor.
1447. The method of item 1363 wherein the agent is an AXOR12 agonist.
1448. The method of item 1363 wherein the agent is an angiotensin Il receptor agonist. 1449. The method of item 1363 wherein the agent is a platelet aggregation inhibitor.
1450. The method of item 1363 wherein the agent is a CB1/CB2 receptor agonist.
1451. The method of item 1363 wherein the agent is a norepinephrine reuptake inhibitor.
1452. The method of item 1363 wherein the agent is a selective serotonin reuptake inhibitor.
1453. The method of item 1363 wherein the agent is a reducing agent.
1454. The method of item 1363 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
1455. The method of item 1363 wherein the agent is isotretinoin.
1456. The method of item 1363 wherein the agent is radicicol.
1457. The method of item 1363 wherein the agent is clobetasol propionate.
1458. The method of item 1363 wherein the agent is homoharringtonine.
1459. The method of item 1363 wherein the agent is trichostatin
A. 1460. The method of item 1363 wherein the agent is brefeldin A.
1461. The method of item 1363 wherein the agent is thapsigargin.
1462. The method of item 1363 wherein the agent is dolastatin 15.
1463. The method of item 1363 wherein the agent is cerivastatin.
1464. The method of item 1363 wherein the agent is jasplakinolide.
1465. The method of item 1363 wherein the agent is herbimycin A.
1466. The method of item 1363 wherein the agent is pirfenidone.
1467. The method of item 1363 wherein the agent is vinorelbine.
1468. The method of item 1363 wherein the agent is 17-DMAG.
1469. The method of item 1363 wherein the agent is tacrolimus.
1470. The method of item 1363 wherein the agent is loteprednol etabonate.
1471. The method of item 1363 wherein the agent is juglone.
1472. The method of item 1363 wherein the agent is prednisolone. 1473. The method of item 1363 wherein the agent is puromycin.
1474. The method of item 1363 wherein the agent is 3-BAABE.
1475. The method of item 1363 wherein the agent is cladribine.
1476. The method of item 1363 wherein the agent is mannose-6- phosphate.
1477. The method of item 1363 wherein the agent is 5- azacytidine.
1478. The method of item 1363 wherein the agent is Ly333531 (ruboxistaurin).
1479. The method of item 1363 wherein the agent is simvastatin.
1480. The method of item 1363, wherein the composition comprises a polymer.
1481. The method of item 1363, wherein the composition comprises a polymeric carrier.
1482. The method of item 1363 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1483. The method of item 1363 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 1484. The method of item 1363 wherein the device has a coating that comprises the anti-scarring agent.
1485. The method of item 1363, wherein the device has a coating that comprises the agent and is disposed on a surface of the sensor.
1486. The method of item 1363, wherein the device has a coating that comprises the agent and directly contacts the sensor.
1487. The method of item 1363, wherein the device has a coating that comprises the agent and indirectly contacts the sensor.
1488. The method of item 1363, wherein the device has a coating that comprises the agent and partially covers the sensor.
1489. The method of item 1363, wherein the device has a coating that comprises the agent and completely covers the sensor.
1490. The method of item 1363, wherein the device has a uniform coating.
1491. The method of item 1363, wherein the device has a nonuniform coating.
1492. The method of item 1363, wherein the device has a discontinuous coating.
1493. The method of item 1363, wherein the device has a patterned coating. 1494. The method of item 1363, wherein the device has a coating with a thickness of 100 μm or less.
1495. The method of item 1363, wherein the device has a coating with a thickness of 10 μm or less.
1496. The method of item 1363, wherein the device has a coating, and the coating adheres to the surface of the sensor upon deployment of the sensor.
1497. The method of item 1363, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
1498. The method of item 1363, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1499. The method of item 1363, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1500. The method of item 1363, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1501. The method of item 1363, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 1502. The method of item 1363, wherein the device has a coating, and wherein the coating further comprises a polymer.
1503. The method of item 1363, wherein the device has a first coating having a first composition and a second coating having a second composition.
1504. The method of item 1363, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
1505. The method of item 1363, wherein the composition comprises a polymer.
1506. The method of item 1363, wherein the composition comprises a polymeric carrier.
1507. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
1508. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
1509. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer. 1510. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
1511. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
1512. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
1513. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
1514. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1515. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1516. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
1517. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer. 1518. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
1519. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
1520. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
1521. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
1522. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
1523. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
1524. The method of item 1363, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1525. The method of item 1363 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer. 1526. The method of item 1363, wherein the device comprises a lubricious coating.
1527. The method of item 1363 wherein the anti-scarring agent is located within pores or holes of the device.
1528. The method of item 1363 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1529. The method of item 1363, wherein the device comprises a second pharmaceutically active agent.
1530. The method of item 1363 wherein the device comprises an anti-inflammatory agent.
1531. The method of item 1363 wherein the device comprises an agent that inhibits infection.
1532. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
1533. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
1534. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
1535. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine. 1536. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5-fluorouracil (5-FU).
1537. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
1538. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
1539. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
1540. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
1541. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
1542. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
1543. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
1544. The method of item 1363 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
1545. The method of item 1363, further comprising an anti- thrombotic agent. 1546. The method of item 1363 wherein the device comprises a visualization agent.
1547. The method of item 1363 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1548. The method of item 1363 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
1549. The method of item 1363 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
1550. The method of item 1363 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
1551. The method of item 1363 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1552. The method of item 1363 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 1553. The method of item 1363 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
1554. The method of item 1363 wherein the device comprises an echogenic material.
1555. The method of item 1363 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
1556. The method of item 1363 wherein the device is sterile.
1557. The method of item 1363 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1558. The method of item 1363 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
1559. The method of item 1363 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
1560. The method of item 1363 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
1561. The method of item 1363 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue. 1562. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1563. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1564. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1565. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1566. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1567. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1568. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1569. The method of item 1363 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti- scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days. 1570. The method of item 1363 wherein the device comprises about 0.01 μg to about 10 μg of the anti-scarring agent.
1571. The method of item 1363 wherein the device comprises about 10 μg to about 10 mg of the anti-scarring agent.
1572. The method of item 1363 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1573. The method of item 1363 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1574. The method of item 1363 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1575. The method of item 1363 wherein a surface of the device comprises less than 0.01 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1576. The method of item 1363 wherein a surface of the device comprises about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1577. The method of item 1363 wherein a surface of the device comprises about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1578. The method of item 1363 wherein a surface of the device comprises about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1579. The method of item 1363 wherein a surface of the device comprises about 250 μg to about 1000 μg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1580. The method of item 1363 wherein a surface of the device comprises ab toouutt 11000000 μμgg ttoo aabboouutt 22550000 μμgg ooff tthhee aannttii--ssccaarrrriirng agent per mm2 of device surface to which the anti-scarring agent is applied.
1581. The method of any one of items 1363-1580 wherein the device is adapted for delivering insulin.
1582. The method of any one of items 1363-1580 wherein the device is adapted for delivering a narcotic.
1583. The method of any one of items 1363-1580 wherein the device is adapted for delivering a chemotherapeutic agent.
1584. The method of any one of items 1363-1580 wherein the device is adapted for delivering an anti-arrhythmic drug.
1585. The method of any one of items 1363-1580 wherein the device is adapted for delivering an anti-spasmotic drug.
1586. The method of any one of items 1363-1580 wherein the device is adapted for delivering an anti-spastic agent.
1587. The method of any one of items 1363-1580 wherein the device is adapted for delivering an antibiotic. 1588. The method of any one of items 1363-1580 wherein the device is adapted for delivering a drug only when changes in the host are detected.
1589. The method of any one of items 1363-1580 wherein the device is adapted for delivering a drug as a continuous slow release.
1590. The method of any one of items 1363-1580 wherein the device is adapted for delivering a drug at prescribed dosages in a pulsatile manner.
1591. The method of any one of items 1363-1580 wherein the device is a programmable drug delivery pump.
1592. The method of any one of items 1363-1580 wherein the device is adapted for intraocularly delivering a drug.
1593. The method of any one of items 1363-1580 wherein the device is adapted for intrathecal^ delivering a drug.
1594. The method of any one of items 1363-1580 wherein the device is adapted for intraperitoneally delivering a drug.
1595. The method of any one of items 1363-1580 wherein the device is adapted for intra-arterially delivering a drug.
1596. The method of any one of items 1363-1580 wherein the device is adapted for intracardiac delivery of a drug.
1597. The method of any one of items 1363-1580 wherein the device is an implantable osmotic pump. 1598. The method of any one of items 1363-1580 wherein the device is an ocular drug delivery pump.
1599. The method of any one of items 1363-1580 wherein the device is metering system.
1600. The method of any one of items 1363-1580 wherein the device is a peristaltic (roller) pump.
1601. The method of any one of items 1363-1580 wherein the device is an electronically driven pump.
1602. The method of any one of items 1363-1580 wherein the device is an elastomeric pump.
1603. The method of any one of items 1363-1580 wherein the device is a spring contraction pump.
1604. The method of any one of items 1363-1580 wherein the device is a gas-driven pump.
1605. The method of any one of items 1363-1580 wherein the device is a hydraulic pump.
1606. The method of any one of items 1363-1580 wherein the device is a piston-dependent pump.
1607. The method of any one of items 1363-1580 wherein the device is a non-piston-dependent pump. 1608. The method of any one of items 1363-1580 wherein the device is a dispensing chamber.
1609. The method of any one of items 1363-1580 wherein the device is an infusion pump.
1610. The method of any one of items 1363-1580 wherein the device is a passive pump.
1611. The method of any one of items 1363-1580 wherein the device is an implantable insulin pump.
1612. The method of item 1611 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense medication through the catheter.
1613. The method of any one of items 1363-1580 wherein the device is an intrathecal drug delivery pump.
1614. The method of item 1613 wherein the device is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord.
1615. The method of item 1613 wherein the device is adapted for delivering a drug to the brain.
1616. The method of item 1613 wherein the device is adapted for intrathecal delivering baclofen. 1617. The method of item 1613 wherein the device further comprises an intraspinal catheter.
1618. The method of item 1613 further comprising a second intrathecal drug delivery pump.
1619. The method of item 1613 wherein the device further comprises a catheter and an electrode.
1620. The method of any one of items 1363-1580 wherein the device is an implantable drug delivery pump for chemotherapy.
1621. The method of item 1620 wherein the device is adapted for delivering 2'-deoxy 5-fluorouridine.
1622. The method of item 1620 wherein the host has a solid tumor, and the device is adapted for infusing a chemotherapeutic agent to the solid tumor.
1623. The method of item 1620 wherein the host has a tumor, and the device is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
1624. The method of item 1620 wherein the host has a hepatic tumor, and the device is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host.
1625. The method of any one of items 1363-1580 wherein the device is a drug delivery pump for treating heart disease. 1626. The method of item 1625 wherein the device is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site.
1627. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a sensor.
1628. The method for implanting a medical device according to item 1627 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
1629. The method for implanting a medical device according to item 1627 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
1630. The method for implanting a medical device according to item 1627 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
1631. The method for implanting a medical device according to item 1627 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
1632. The method for implanting a medical device according to item 1627 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
1633. The method for implanting a medical device according to item 1627 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
1634. The method of item 1627 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
1635. The method of item 1627 wherein the anti-fibrotic agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
1636. The method of item 1627 wherein the anti-fibrotic agent is mevastatin (a fibrinogen antagonist).
1637. The method of item 1627 wherein the anti-fibrotic agent is vincamine (a microtubule inhibitor). 1638. The method of item 1627 wherein the anti-fibrotic agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
1639. The method of item 1627 wherein the anti-fibrotic agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
1640. The method of item 1627 wherein the anti-fibrotic agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L- 165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
1641. The method of item 1627 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
1642. The method of item 1627 wherein the anti-fibrotic agent is a tumor necrosis factor antagonist.
1643. The method of item 1627 wherein the anti-fibrotic agent is herbimycin A (a tyrosine kinase inhibitor).
1644. The method of item 1627 wherein the anti-fibrotic agent is erucylphosphocholine.
1645. The method of item 1627 wherein the anti-fibrotic agent is alphastatin. 1646. The method of item 1627 wherein the anti-fibrotic agent is etanercept.
1647. The method of item 1627 wherein the anti-fibrotic agent is humicade.
1648. The method of item 1627 wherein the anti-fibrotic agent is gefitinib.
1649. The method of item 1627 wherein the anti-fibrotic agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
1650. The method of item 1627 wherein the anti-fibrotic agent is an alpha adrenergic receptor antagonist.
1651. The method of item 1627 wherein the anti-fibrotic agent is an anti-psychotic compound. 1652. The method of item 1627 wherein the anti-fibrotic agent is a CaM kinase Il inhibitor.
1653. The method of item 1627 wherein the anti-fibrotic agent is a G protein agonist.
1654. The method of item 1627 wherein the anti-fibrotic agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
1655. The method of item 1627 wherein the anti-fibrotic agent is an anti-microbial agent.
1656. The method of item 1627 wherein the anti-fibrotic agent is a DNA topoisomerase inhibitor selected from the group consisting of β- lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
1657. The method of item 1627 wherein the anti-fibrotic agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
1658. The method of item 1627 wherein the anti-fibrotic agent is a D2 dopamine receptor antagonist.
1659. The method of item 1627 wherein the anti-fibrotic agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor. 1660. The method of item 1627 wherein the anti-fibrotic agent is a dopamine antagonist.
1661. The method of item 1627 wherein the anti-fibrotic agent is an anesthetic compound.
1662. The method of item 1627 wherein the anti-fibrotic agent is a clotting factor.
1663. The method of item 1627 wherein the anti-fibrotic agent is a lysyl hydrolase inhibitor.
1664. The method of item 1627 wherein the anti-fibrotic agent is a muscarinic receptor inhibitor.
1665. The method of item 1627 wherein the anti-fibrotic agent is a superoxide anion generator.
1666. The method of item 1627 wherein the anti-fibrotic agent is a steroid.
1667. The method of item 1627 wherein the anti-fibrotic agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
1668. The method of item 1627 wherein the anti-fibrotic agent is a diuretic. 1669. The method of item 1627 wherein the anti-fibrotic agent is an anti-coagulant.
1670. The method of item 1627 wherein the anti-fibrotic agent is a cyclic GMP agonist.
1671. The method of item 1627 wherein the anti-fibrotic agent is an adenylate cyclase agonist.
1672. The method of item 1627 wherein the anti-fibrotic agent is an antioxidant.
1673. The method of item 1627 wherein the anti-fibrotic agent is a nitric oxide synthase inhibitor.
1674. The method of item 1627 wherein the anti-fibrotic agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
1675. The method of item 1627 wherein the anti-fibrotic agent is a DNA synthesis inhibitor.
1676. The method of item 1627 wherein the anti-fibrotic agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
1677. The method of item 1627 wherein the anti-fibrotic agent is a DNA methylation inhibitor. 1678. The method of item 1627 wherein the anti-fibrotic agent is a NSAID agent.
1679. The method of item 1627 wherein the anti-fibrotic agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
1680. The method of item 1627 wherein the anti-fibrotic agent is an MEK1/MEK 2 inhibitor.
1681. The method of item 1627 wherein the anti-fibrotic agent is a NO synthase inhibitor.
1682. The method of item 1627 wherein the anti-fibrotic agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-
2) and an analogue or derivative thereof.
1683. The method of item 1627 wherein the anti-fibrotic agent is an ACE inhibitor.
1684. The method of item 1627 wherein the anti-fibrotic agent is a glycosylation inhibitor.
1685. The method of item 1627 wherein the anti-fibrotic agent is an intracellular calcium influx inhibitor.
1686. The method of item 1627 wherein the anti-fibrotic agent is an anti-emetic agent.
1687. The method of item 1627 wherein the anti-fibrotic agent is an acetylcholinesterase inhibitor. 1688. The method of item 1627 wherein the anti-fibrotic agent is an ALK-5 receptor antagonist.
1689. The method of item 1627 wherein the anti-fibrotic agent is a RAR/RXT antagonist.
1690. The method of item 1627 wherein the anti-fibrotic agent is an elF-2a inhibitor.
1691. The method of item 1627 wherein the anti-fibrotic agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
1692. The method of item 1627 wherein the anti-fibrotic agent is an estrogen agonist.
1693. The method of item 1627 wherein the anti-fibrotic agent is a serotonin receptor inhibitor.
1694. The method of item 1627 wherein the anti-fibrotic agent is an anti-thrombotic agent.
1695. The method of item 1627 wherein the anti-fibrotic agent is a tryptase inhibitor.
1696. The method of item 1627 wherein the anti-fibrotic agent is a pesticide.
1697. The method of item 1627 wherein the anti-fibrotic agent is a bone mineralization promotor. 1698. The method of item 1627 wherein the anti-fibrotic agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
1699. The method of item 1627 wherein the anti-fibrotic agent is an anti-inflammatory compound.
1700. The method of item 1627 wherein the anti-fibrotic agent is a DNA methylation promotor.
1701. The method of item 1627 wherein the anti-fibrotic agent is an anti-spasmodic agent.
1702. The method of item 1627 wherein the anti-fibrotic agent is a protein synthesis inhibitor.
1703. The method of item 1627 wherein the anti-fibrotic agent is an α-glucosidase inhibitor.
1704. The method of item 1627 wherein the anti-fibrotic agent is a calcium channel blocker.
1705. The method of item 1627 wherein the anti-fibrotic agent is a pyruvate dehydrogenase activator.
1706. The method of item 1627 wherein the anti-fibrotic agent is a prostaglandin inhibitor.
1707. The method of item 1627 wherein the anti-fibrotic agent is a sodium channel inhibitor. 1708. The method of item 1627 wherein the anti-fibrotic agent is a serine protease inhibitor.
1709. The method of item 1627 wherein the anti-fibrotic agent is an intracellular calcium flux inhibitor.
1710. The method of item 1627 wherein the anti-fibrotic agent is a JAK2 inhibitor.
1711. The method of item 1627 wherein the anti-fibrotic agent is an androgen inhibitor.
1712. The method of item 1627 wherein the anti-fibrotic agent is an aromatase inhibitor.
1713. The method of item 1627 wherein the anti-fibrotic agent is an anti-viral agent.
1714. The method of item 1627 wherein the anti-fibrotic agent is a 5-HT inhibitor.
1715. The method of item 1627 wherein the anti-fibrotic agent is an FXR antagonist.
1716. The method of item 1627 wherein the agent is an actin polymerization and stabilization promotor.
1717. The method of item 1627 wherein the anti-fibrotic agent is an AXOR12 agonist. 1718. The method of item 1627 wherein the anti-fibrotic agent is an angiotensin Il receptor agonist.
1719. The method of item 1627 wherein the anti-fibrotic agent is a platelet aggregation inhibitor.
1720. The method of item 1627 wherein the anti-fibrotic agent is a CB1/CB2 receptor agonist.
1721. The method of item 1627 wherein the anti-fibrotic agent is a norepinephrine reuptake inhibitor.
1722. The method of item 1627 wherein the anti-fibrotic agent is a selective serotonin reuptake inhibitor.
1723. The method of item 1627 wherein the anti-fibrotic agent is a reducing agent.
1724. The method of item 1627 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
1725. The method of item 1627 wherein the anti-fibrotic agent is isotretinoin.
1726. The method of item 1627 wherein the anti-fibrotic agent is radicicol.
1727. The method of item 1627 wherein the agent is clobetasol propionate. 1728. The method of item 1627 wherein the anti-fibrotic agent is homoharringtonine.
1729. The method of item 1627 wherein the anti-fibrotic agent is trichostatin A.
1730. The method of item 1627 wherein the anti-fibrotic agent is brefeldin A.
1731. The method of item 1627 wherein the anti-fibrotic agent is thapsigargin.
1732. The method of item 1627 wherein the anti-fibrotic agent is dolastatin 15.
1733. The method of item 1627 wherein the anti-fibrotic agent is cerivastatin.
1734. The method of item 1627 wherein the anti-fibrotic agent is jasplakinolide.
1735. The method of item 1627 wherein the anti-fibrotic agent is herbimycin A.
1736. The method of item 1627 wherein the anti-fibrotic agent is pirfenidone.
1737. The method of item 1627 wherein the anti-fibrotic agent is vinorelbine. 1738. The method of item 1627 wherein the anti-fibrotic agent is 17-DMAG.
1739. The method of item 1627 wherein the anti-fibrotic agent is tacrolimus.
1740. The method of item 1627 wherein the anti-fibrotic agent is loteprednol etabonate.
1741. The method of item 1627 wherein the anti-fibrotic agent is juglone.
1742. The method of item 1627 wherein the anti-fibrotic agent is prednisolone.
1743. The method of item 1627 wherein the anti-fibrotic agent is puromycin.
1744. The method of item 1627 wherein the anti-fibrotic agent is 3-BAABE.
1745. The method of item 1627 wherein the anti-fibrotic agent is cladribine.
1746. The method of item 1627 wherein the anti-fibrotic agent is mannose-6-phosphate.
1747. The method of item 1627 wherein the anti-fibrotic agent is 5-azacytidine. 1748. The method of item 1627 wherein the anti-fibrotic agent is Ly333531 (ruboxistaurin).
1749. The method of item 1627 wherein the anti-fibrotic agent is simvastatin.
1750. The method of item 1627 wherein the anti-infective agent is an anthracycline.
1751. The method of item 1627 wherein the anti-infective agent is doxorubicin.
1752. The method of item 1627 wherein the anti-infective agent is mitoxantrone.
1753. The method of item 1627 wherein the anti-infective agent is a fluoropyrimidine.
1754. The method of item 1627 wherein the anti-infective agent is 5-fluorouracil (5-FU).
1755. The method of item 1627 wherein the anti-infective agent is a folic acid antagonist.
1756. The method of item 1627 wherein the anti-infective agent is methotrexate.
1757. The method of item 1627 wherein the anti-infective agent is a podophylotoxin. 1758. The method of item 1627 wherein the anti-infective agent is etoposide.
1759. The method of item 1627 wherein the anti-infective agent is camptothecin.
1760. The method of item 1627 wherein the anti-infective agent is a hydroxyurea.
1761. The method of item 1627 wherein the anti-infective agent is a platinum complex.
1762. The method of item 1627 wherein the anti-infective agent is cisplatin.
1763. The method of item 1627 wherein the composition comprises an anti-thrombotic agent.
1764. The method of item 1627 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
1765. The method of item 1627 wherein the polymer is formed from reactants comprising protein.
1766. The method of item 1627 wherein the polymer is formed from reactants comprising carbohydrate.
1767. The method of item 1627 wherein the polymer is formed from reactants comprising biodegradable polymer. 1768. The method of item 1627 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
1769. The method of item 1627 wherein the polymer is formed from reactants comprising collagen.
1770. The method of item 1627 wherein the polymer is formed from reactants comprising methylated collagen.
1771. The method of item 1627 wherein the polymer is formed from reactants comprising fibrinogen.
1772. The method of item 1627 wherein the polymer is formed from reactants comprising thrombin.
1773. The method of item 1627 wherein the polymer is formed from reactants comprising blood plasma.
1774. The method of item 1627 wherein the polymer is formed from reactants comprising calcium salt.
1775. The method of item 1627 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
1776. The method of item 1627 wherein the polymer is formed from reactants comprising fibrinogen analog.
1777. The method of item 1627 wherein the polymer is formed from reactants comprising albumin. 1778. The method of item 1627 wherein the polymer is formed from reactants comprising plasminogen.
1779. The method of item 1627 wherein the polymer is formed from reactants comprising von Willebrands factor.
1780. The method of item 1627 wherein the polymer is formed from reactants comprising Factor VIII.
1781. The method of item 1627 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
1782. The method of item 1627 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
1783. The method of item 1627 wherein the polymer is formed from reactants comprising telopeptide collagen.
1784. The method of item 1627 wherein the polymer is formed from reactants comprising crosslinked collagen.
1785. The method of item 1627 wherein the polymer is formed from reactants comprising aprotinin.
1786. The method of item 1627 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
1787. The method of item 1627 wherein the polymer is formed from reactants comprising gelatin. 1788. The method of item 1627 wherein the polymer is formed from reactants comprising protein conjugates.
1789. The method of item 1627 wherein the polymer is formed from reactants comprising gelatin conjugates.
1790. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic polymer.
1791. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
1792. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
1793. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
1794. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
1795. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
1796. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound. 1797. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
1798. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
1799. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
1800. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group. "
1801. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
1802. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
1803. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
1804. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 1805. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
1806. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
1807. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
1808. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
1809. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
1810. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
1811. The method of item 1627 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
1812. The method of item 1627 wherein the polymer is formed from reactants comprising polylysine. 1813. The method of item 1627 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
1814. The method of item 1627 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
1815. The method of item 1627 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
1816. The method of item 1627 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
1817. The method of item 1627 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
1818. The method of item 1627 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
1819. The method of item 1627 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
1820. The method of item 1627 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 1821. The method of item 1627 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
1822. The method of item 1627 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
1823. The method of item 1627 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
1824. The method of item 1627 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
1825. The method of item 1627 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
1826. The method of item 1627 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
1827. The method of item 1627 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
1828. The method of item 1627 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups. 1829. The method of item 1627 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
1830. The method of item 1627 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
1831. The method of item 1627 wherein the polymer is formed from reactants comprising hyaluronic acid.
1832. The method of item 1627 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
1833. The method of item 1627 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
1834. The method of item 1627 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
1835. The method of item 1627 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups. 1836. The method of item 1627 wherein the composition comprises a colorant.
1837. The method of item 1627 wherein the composition is sterile.
1838. The method of any one of items 1627-1837 wherein the sensor is a blood or tissue glucose monitor.
1839. The method of any one of items 1627-1837 wherein the sensor is an electrolyte sensor.
1840. The method of any one of items 1627-1837 wherein the sensor is a blood constituent sensor.
1841. The method of any one of items 1627-1837 wherein the sensor is a temperature sensor.
1842. The method of any one of items 1627-1837 wherein the sensor is a pH sensor.
1843. The method of any one of items 1627-1837 wherein the sensor is an optical sensor.
1844. The method of any one of items 1627-1837 wherein the sensor is an amperometric sensor.
1845. The method of any one of items 1627-1837 wherein the sensor is a pressure or stress sensor. 1846. The method of any one of items 1627-1837 wherein the sensor is a biosensor.
1847. The method of any one of items 1627-1837 wherein the sensor is a sensing transponder.
1848. The method of any one of items 1627-1837 wherein the sensor is a strain sensor.
1849. The method of any one of items 1627-1837 wherein the sensor is a magnetoresistive sensor.
1850. The method of any one of items 1627-1837 wherein the sensor is a cardiac sensor.
1851. The method of any one of items 1627-1837 wherein the sensor is a respiratory sensor.
1852. The method of any one of items 1627-1837 wherein the sensor is an auditory sensor.
1853. The method of any one of items 1627-1837 wherein the sensor is a metabolite sensor.
1854. The method of any one of items 1627-1837 wherein the sensor detects mechanical changes.
1855. The method of any one of items 1627-1837 wherein the sensor detects physical changes. 1856. The method of any one of items 1627-1837 wherein the sensor detects electrochemical changes.
1857. The method of any one of items 1627-1837 wherein the sensor detects magnetic changes.
1858. The method of any one of items 1627-1837 wherein the sensor detects acceleration changes.
1859. The method of any one of items 1627-1837 wherein the sensor detects ionizing radiation changes.
1860. The method of any one of items 1627-1837 wherein the sensor detects acoustic wave changes.
1861. The method of any one of items 1627-1837 wherein the sensor detects chemical changes.
1862. The method of any one of items 1627-1837 wherein the sensor detects drug concentration changes.
1863. The method of any one of items 1627-1837 wherein the sensor detects hormone changes.
1864. The method of any one of items 1627-1837 wherein the sensor detects barometric changes.
1865. The method of item 1838 wherein the device is deliverable to the vascular system transluminal^ using a catheter on a stent platform. 1866. The method of item 1838 wherein the device is composed of glucose sensitive living cells that monitor blood glucose levels and produce a detectable electrical or optical signal in response to changes in glucose concentrations.
1867. The method of item 1838 wherein the device is an electrode composed of an analyte responsive enzyme.
1868. The method of item 1838 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates an insulin pump to supply insulin.
1869. The method of item 1838 wherein the device is a closed loop insulin delivery system that comprises a sensing means that detects the host's blood glucose level and stimulates the pancreas to supply insulin.
1870. The method of item 1845 wherein the device monitors blood pressure.
1871. The method of item 1845 wherein the device monitors fluid flow.
1872. The method of item 1845 wherein the device monitors pressure within an aneurysm sac.
1873. The method of item 1845 wherein the device monitors intracranial pressure.
1874. The method of item 1845 wherein the device monitors mechanical pressure associated with a bone fracture. 1875. The method of item 1845 wherein the device monitors barometric pressure.
1876. The method of item 1845 wherein the device monitors eye tremors.
1877. The method of item 1845 wherein the device monitors the depth of a corneal implant.
1878. The method of item 1845 wherein the device monitors intraocular pressure.
1879. The method of item 1845 wherein the device is a passive sensor with an inductor-capacitor circuit.
1880. The method of item 1845 wherein the device is a self- powered strain sensing system.
1881. The method of item 1845 wherein the sensor comprises a lead, a sensor module, a sensor circuit and means for providing voltage.
1882. The method of item 1850 wherein the device monitors cardiac output.
1883. The method of item 1850 wherein the device monitors ejection fraction.
1884. The method of item 1850 wherein the device monitors blood pressure in a heart chamber. 1885. The method of item 1850 wherein the device monitors ventricular wall motions.
1886. The method of item 1850 wherein the device monitors blood flow to a transplanted organ.
1887. The method of item 1850 wherein the device monitors heart rate.
1888. The method of item 1851 wherein the device monitors pulmonary functions.
1889. The method of item 1852 wherein the device is adapted for delivering an electrical signal to an implantable electromechanical transducer that acts on the middle or inner ear.
1890. The method of item 1852 wherein the device generates an electrical audio signal.
1891. The method of item 1852 wherein the device is a capacitive sensor that is coupled to a vibrating auditory element.
1892. The method of item 1852 wherein the device is an electromagnetic sensor.
1893. The method of any one of items 1845 or 1853 wherein the device emits a source of radiation directed towards blood to interact with a plurality of detectors that provide an output signal.
1894. The method of any one of items 1845 or 1853 wherein the device is a biosensing transponder composed of a dye that has optical properties that change in response to changes in the environment, a photosensor to sense the optical changes, and a transponder for transmitting data to a remote reader.
1895. The method of any one of items 1845 or 1853 wherein the device is a monolithic bioelectronic device for detecting at least one analyte within the host.
1896. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a pump.
1897. The method for implanting a medical device according to item 1896 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
1898. The method for implanting a medical device according to item 1896 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
1899. The method for implanting a medical device according to item 1896 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host. 1900. The method for implanting a medical device according to item 1896 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
1901. The method for implanting a medical device according to item 1896 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
1902. The method for implanting a medical device according to item 1896 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
1903. The method of item 1896 wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP
74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
1904. The method of item 1896 wherein the anti-fibrotic agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
1905. The method of item 1896 wherein the anti-fibrotic agent is mevastatin (a fibrinogen antagonist).
1906. The method of item 1896 wherein the anti-fibrotic agent is vincamine (a microtubule inhibitor). 1907. The method of item 1896 wherein the anti-fibrotic agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
1908. The method of item 1896 wherein the anti-fibrotic agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
1909. The method of item 1896 wherein the anti-fibrotic agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof.
1910. The method of item 1896 wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
1911. The method of item 1896 wherein the anti-fibrotic agent is a tumor necrosis factor antagonist.
1912. The method of item 1896 wherein the anti-fibrotic agent is herbimycin A (a tyrosine kinase inhibitor).
1913. The method of item 1896 wherein the anti-fibrotic agent is erucylphosphocholine.
1914. The method of item 1896 wherein the anti-fibrotic agent is alphastatin. 1915. The method of item 1896 wherein the anti-fibrotic agent is etanercept.
1916. The method of item 1896 wherein the anti-fibrotic agent is humicade.
1917. The method of item 1896 wherein the anti-fibrotic agent is gefitinib.
1918. The method of item 1896 wherein the anti-fibrotic agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
1919. The method of item 1896 wherein the anti-fibrotic agent is an alpha adrenergic receptor antagonist.
1920. The method of item 1896 wherein the anti-fibrotic agent is an anti-psychotic compound. 1921. The method of item 1896 wherein the anti-fibrotic agent is a CaM kinase Il inhibitor.
1922. The method of item 1896 wherein the anti-fibrotic agent is a G protein agonist.
1923. The method of item 1896 wherein the anti-fibrotic agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
1924. The method of item 1896 wherein the anti-fibrotic agent is an anti-microbial agent.
1925. The method of item 1896 wherein the anti-fibrotic agent is a DNA topoisomerase inhibitor selected from the group consisting of β- lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
1926. The method of item 1896 wherein the anti-fibrotic agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
1927. The method of item 1896 wherein the anti-fibrotic agent is a D2 dopamine receptor antagonist.
1928. The method of item 1896 wherein the anti-fibrotic agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor. 1929. The method of item 1896 wherein the anti-fibrotic agent is a dopamine antagonist.
1930. The method of item 1896 wherein the anti-fibrotic agent is an anesthetic compound.
1931. The method of item 1896 wherein the anti-fibrotic agent is a clotting factor.
1932. The method of item 1896 wherein the anti-fibrotic agent is a lysyl hydrolase inhibitor.
1933. The method of item 1896 wherein the anti-fibrotic agent is a muscarinic receptor inhibitor.
1934. The method of item 1896 wherein the anti-fibrotic agent is a superoxide anion generator.
1935. The method of item 1896 wherein the anti-fibrotic agent is a steroid.
1936. The method of item 1896 wherein the anti-fibrotic agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
1937. The method of item 1896 wherein the anti-fibrotic agent is a diuretic. 1938. The method of item 1896 wherein the anti-fibrotic agent is an anti-coagulant.
1939. The method of item 1896 wherein the anti-fibrotic agent is a cyclic GMP agonist.
1940. The method of item 1896 wherein the anti-fibrotic agent is an adenylate cyclase agonist.
1941. The method of item 1896 wherein the anti-fibrotic agent is an antioxidant.
1942. The method of item 1896 wherein the anti-fibrotic agent is a nitric oxide synthase inhibitor.
1943. The method of item 1896 wherein the anti-fibrotic agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
1944. The method of item 1896 wherein the anti-fibrotic agent is a DNA synthesis inhibitor.
1945. The method of item 1896 wherein the anti-fibrotic agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
1946. The method of item 1896 wherein the anti-fibrotic agent is a DNA methylation inhibitor. 1947. The method of item 1896 wherein the anti-fibrotic agent is a NSAID agent.
1948. The method of item 1896 wherein the anti-fibrotic agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
1949. The method of item 1896 wherein the anti-fibrotic agent is an MEK1/MEK 2 inhibitor.
1950. The method of item 1896 wherein the anti-fibrotic agent is a NO synthase inhibitor.
1951. The method of item 1896 wherein the anti-fibrotic agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-
2) and an analogue or derivative thereof.
1952. The method of item 1896 wherein the anti-fibrotic agent is an ACE inhibitor.
1953. The method of item 1896 wherein the anti-fibrotic agent is a glycosylation inhibitor.
1954. The method of item 1896 wherein the anti-fibrotic agent is an intracellular calcium influx inhibitor.
1955. The method of item 1896 wherein the anti-fibrotic agent is an anti-emetic agent.
1956. The method of item 1896 wherein the anti-fibrotic agent is an acetylcholinesterase inhibitor. 1957. The method of item 1896 wherein the anti-fibrotic agent is an ALK-5 receptor antagonist.
1958. The method of item 1896 wherein the anti-fibrotic agent is a RAR/RXT antagonist.
1959. The method of item 1896 wherein the anti-fibrotic agent is an elF-2a inhibitor.
1960. The method of item 1896 wherein the anti-fibrotic agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
1961. The method of item 1896 wherein the anti-fibrotic agent is an estrogen agonist.
1962. The method of item 1896 wherein the anti-fibrotic agent is a serotonin receptor inhibitor.
1963. The method of item 1896 wherein the anti-fibrotic agent is an anti-thrombotic agent.
1964. The method of item 1896 wherein the anti-fibrotic agent is a tryptase inhibitor.
1965. The method of item 1896 wherein the anti-fibrotic agent is a pesticide.
1966. The method of item 1896 wherein the anti-fibrotic agent is a bone mineralization promotor. 1967. The method of item 1896 wherein the anti-fibrotic agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
1968. The method of item 1896 wherein the anti-fibrotic agent is an anti-inflammatory compound.
1969. The method of item 1896 wherein the anti-fibrotic agent is a DNA methylation promotor.
1970. The method of item 1896 wherein the anti-fibrotic agent is an anti-spasmodic agent.
1971. The method of item 1896 wherein the anti-fibrotic agent is a protein synthesis inhibitor.
1972. The method of item 1896 wherein the anti-fibrotic agent is an α-glucosidase inhibitor.
1973. The method of item 1896 wherein the anti-fibrotic agent is a calcium channel blocker.
1974. The method of item 1896 wherein the anti-fibrotic agent is a pyruvate dehydrogenase activator.
1975. The method of item 1896 wherein the anti-fibrotic agent is a prostaglandin inhibitor.
1976. The method of item 1896 wherein the anti-fibrotic agent is a sodium channel inhibitor. 1977. The method of item 1896 wherein the anti-fibrotic agent is a serine protease inhibitor.
1978. The method of item 1896 wherein the anti-fibrotic agent is an intracellular calcium flux inhibitor.
1979. The method of item 1896 wherein the anti-fibrotic agent is a JAK2 inhibitor.
1980. The method of item 1896 wherein the anti-fibrotic agent is an androgen inhibitor.
1981. The method of item 1896 wherein the anti-fibrotic agent is an aromatase inhibitor.
1982. The method of item 1896 wherein the anti-fibrotic agent is an anti-viral agent.
1983. The method of item 1896 wherein the anti-fibrotic agent is a 5-HT inhibitor.
1984. The method of item 1896 wherein the anti-fibrotic agent is an FXR antagonist.
1985. The method of item 1896 wherein the agent is an actin polymerization and stabilization promotor.
1986. The method of item 1896 wherein the anti-fibrotic agent is an AXOR12 agonist. 1987. The method of item 1896 wherein the anti-fibrotic agent is an angiotensin Il receptor agonist.
1988. The method of item 1896 wherein the anti-fibrotic agent is a platelet aggregation inhibitor.
1989. The method of item 1896 wherein the anti-fibrotic agent is a CB1/CB2 receptor agonist.
1990. The method of item 1896 wherein the anti-fibrotic agent is a norepinephrine reuptake inhibitor.
1991. The method of item 1896 wherein the anti-fibrotic agent is a selective serotonin reuptake inhibitor.
1992. The method of item 1896 wherein the anti-fibrotic agent is a reducing agent.
1993. The method of item 1896 wherein the agent is a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
1994. The method of item 1896 wherein the anti-fibrotic agent is isotretinoin.
1995. The method of item 1896 wherein the anti-fibrotic agent is radicicol.
1996. The method of item 1896 wherein the agent is clobetasol propionate. 1997. The method of item 1896 wherein the anti-fibrotic agent is homoharringtonine.
1998. The method of item 1896 wherein the anti-fibrotic agent is trichostatin A.
1999. The method of item 1896 wherein the anti-fibrotic agent is brefeldin A.
2000. The method of item 1896 wherein the anti-fibrotic agent is thapsigargin.
2001. The method of item 1896 wherein the anti-fibrotic agent is dolastatin 15.
2002. The method of item 1896 wherein the anti-fibrotic agent is cerivastatin.
2003. The method of item 1896 wherein the anti-fibrotic agent is jasplakinolide.
2004. The method of item 1896 wherein the anti-fibrotic agent is herbimycin A.
2005. The method of item 1896 wherein the anti-fibrotic agent is pirfenidone.
2006. The method of item 1896 wherein the anti-fibrotic agent is vinorelbine. 2007. The method of item 1896 wherein the anti-fibrotic agent is 17-DMAG.
2008. The method of item 1896 wherein the anti-fibrotic agent is tacrolimus.
2009. The method of item 1896 wherein the anti-fibrotic agent is loteprednol etabonate.
2010. The method of item 1896 wherein the anti-fibrotic agent is juglone.
2011. The method of item 1896 wherein the anti-fibrotic agent is prednisolone.
2012. The method of item 1896 wherein the anti-fibrotic agent is puromycin.
2013. The method of item 1896 wherein the anti-fibrotic agent is 3-BAABE.
2014. The method of item 1896 wherein the anti-fibrotic agent is cladribine.
2015. The method of item 1896 wherein the anti-fibrotic agent is mannose-6-phosphate.
2016. The method of item 1896 wherein the anti-fibrotic agent is 5-azacytidine. 2017. The method of item 1896 wherein the anti-fibrotic agent is Ly333531 (ruboxistaurin).
2018. The method of item 1896 wherein the anti-fibrotic agent is simvastatin.
2019. The method of item 1896 wherein the anti-infective agent is an anthracycline.
2020. The method of item 1896 wherein the anti-infective agent is doxorubicin.
2021. The method of item 1896 wherein the anti-infective agent is mitoxantrone.
2022. The method of item 1896 wherein the anti-infective agent is a fluoropyrimidine.
2023. The method of item 1896 wherein the anti-infective agent is 5-fluorouracil (5-FU).
2024. The method of item 1896 wherein the anti-infective agent is a folic acid antagonist.
2025. The method of item 1896 wherein the anti-infective agent is methotrexate.
2026. The method of item 1896 wherein the anti-infective agent is a podophylotoxin. 2027. The method of item 1896 wherein the anti-infective agent is etoposide.
2028. The method of item 1896 wherein the anti-infective agent is camptothecin.
2029. The method of item 1896 wherein the anti-infective agent is a hydroxyurea.
2030. The method of item 1896 wherein the anti-infective agent is a platinum complex.
2031. The method of item 1896 wherein the anti-infective agent is cisplatin.
2032. The method of item 1896 wherein the composition comprises an anti-thrombotic agent.
2033. The method of item 1896 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
2034. The method of item 1896 wherein the polymer is formed from reactants comprising protein.
2035. The method of item 1896 wherein the polymer is formed from reactants comprising carbohydrate.
2036. The method of item 1896 wherein the polymer is formed from reactants comprising biodegradable polymer. 2037. The method of item 1896 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
2038. The method of item 1896 wherein the polymer is formed from reactants comprising collagen.
2039. The method of item 1896 wherein the polymer is formed from reactants comprising methylated collagen.
2040. The method of item 1896 wherein the polymer is formed from reactants comprising fibrinogen.
2041. The method of item 1896 wherein the polymer is formed from reactants comprising thrombin.
2042. The method of item 1896 wherein the polymer is formed from reactants comprising blood plasma.
2043. The method of item 1896 wherein the polymer is formed from reactants comprising calcium salt.
2044. The method of item 1896 wherein the polymer is formed from reactants comprising an antifibronolytic agent.
2045. The method of item 1896 wherein the polymer is formed from reactants comprising fibrinogen analog.
2046. The method of item 1896 wherein the polymer is formed from reactants comprising albumin. 2047. The method of item 1896 wherein the polymer is formed from reactants comprising plasminogen.
2048. The method of item 1896 wherein the polymer is formed from reactants comprising von Willebrands factor.
2049. The method of item 1896 wherein the polymer is formed from reactants comprising Factor VIII.
2050. The method of item 1896 wherein the polymer is formed from reactants comprising hypoallergenic collagen.
2051. The method of item 1896 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
2052. The method of item 1896 wherein the polymer is formed from reactants comprising telopeptide collagen.
2053. The method of item 1896 wherein the polymer is formed from reactants comprising crosslinked collagen.
2054. The method of item 1896 wherein the polymer is formed from reactants comprising aprotinin.
2055. The method of item 1896 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
2056. The method of item 1896 wherein the polymer is formed from reactants comprising gelatin. 2057. The method of item 1896 wherein the polymer is formed from reactants comprising protein conjugates.
2058. The method of item 1896 wherein the polymer is formed from reactants comprising gelatin conjugates.
2059. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic polymer.
2060. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
2061. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
2062. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
2063. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
2064. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
2065. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound. 2066. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
2067. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
2068. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
2069. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl- oxygen-succinimidyl group.
2070. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
2071. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
2072. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
2073. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 2074. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
2075. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
2076. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
2077. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
2078. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
2079. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
2080. The method of item 1896 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
2081. The method of item 1896 wherein the polymer is formed from reactants comprising polylysine. 2082. The method of item 1896 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
2083. The method of item 1896 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
2084. The method of item 1896 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
2085. The method of item 1896 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
2086. The method of item 1896 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
2087. The method of item 1896 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
2088. The method of item 1896 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
2089. The method of item 1896 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 2090. The method of item 1896 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
2091. The method of item 1896 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
2092. The method of item 1896 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
2093. The method of item 1896 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
2094. The method of item 1896 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
2095. The method of item 1896 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
2096. The method of item 1896 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
2097. The method of item 1896 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups. 2098. The method of item 1896 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
2099. The method of item 1896 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
2100. The method of item 1896 wherein the polymer is formed from reactants comprising hyaluronic acid.
2101. The method of item 1896 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
2102. The method of item 1896 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
2103. The method of item 1896 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
2104. The method of item 1896 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups. 2105. The method of item 1896 wherein the composition comprises a colorant.
2106. The method of item 1896 wherein the composition is sterile.
2107. The method of any one of items 1896-2106 wherein the device is adapted for delivering insulin.
2108. The method of any one of items 1896-2106 wherein the device is adapted for delivering a narcotic.
2109. The method of any one of items 1896-2106 wherein the device is adapted for delivering a chemotherapeutic agent.
2110. The method of any one of items 1896-2106 wherein the device is adapted for delivering an anti-arrhythmic drug.
2111. The method of any one of items 1896-2106 wherein the device is adapted for delivering an anti-spasmotic drug.
2112. The method of any one of items 1896-2106 wherein the device is adapted for delivering an anti-spastic agent.
2113. The method of any one of items 1896-2106 wherein the device is adapted for delivering an antibiotic.
2114. The method of any one of items 1896-2106 wherein the device is adapted for delivering a drug only when changes in the host are detected. 2115. The method of any one of items 1896-2106 wherein the device is adapted for delivering a drug as a continuous slow release.
2116. The method of any one of items 1896-2106 wherein the device is adapted for delivering a drug at prescribed dosages in a pulsatile manner.
2117. The method of any one of items 1896-2106 wherein the device is a programmable drug delivery pump.
2118. The method of any one of items 1896-2106 wherein the device is adapted for intraocularly delivering a drug.
2119. The method of any one of items 1896-2106 wherein the device is adapted for intrathecally delivering a drug.
2120. The method of any one of items 1896-2106 wherein the device is adapted for intraperitoneally delivering a drug.
2121. The method of any one of items 1896-2106 wherein the device is adapted for intra-arterially delivering a drug.
2122. The method of any one of items 1896-2106 wherein the device is adapted for intracardiac delivery of a drug.
2123. The method of any one of items 1896-2106 wherein the device is an implantable osmotic pump.
2124. The method of any one of items 1896-2106 wherein the device is an ocular drug delivery pump. 2125. The method of any one of items 1896-2106 wherein the device is metering system.
2126. The method of any one of items 1896-2106 wherein the device is a peristaltic (roller) pump.
2127. The method of any one of items 1896-2106 wherein the device is an electronically driven pump.
2128. The method of any one of items 1896-2106 wherein the device is an elastomeric pump.
2129. The method of any one of items 1896-2106 wherein the device is a spring contraction pump.
2130. The method of any one of items 1896-2106 wherein the device is a gas-driven pump.
2131. The method of any one of items 1896-2106 wherein the device is a hydraulic pump.
2132. The method of any one of items 1896-2106 wherein the device is a piston-dependent pump.
2133. The method of any one of items 1896-2106 wherein the device is a non-piston-dependent pump.
2134. The method of any one of items 1896-2106 wherein the device is a dispensing chamber. 2135. The method of any one of items 1896-2106 wherein the device is an infusion pump.
2136. The method of any one of items 1896-2106 wherein the device is a passive pump.
2137. The method of any one of items 1896-2106 wherein the device is an implantable insulin pump.
2138. The method of item 2137 further comprising a single channel catheter with a sensor implanted in a vessel that transmits blood chemistry to the implantable insulin pump to dispense medication through the catheter.
2139. The method of any one of items 1896-2106 wherein the device is an intrathecal drug delivery pump.
2140. The method of item 2139 wherein the device is adapted for delivering pain medication directly into the cerebrospinal fluid of the intrathecal space surrounding the spinal cord.
2141. The method of item 2139 wherein the device is adapted for delivering a drug to the brain.
2142. The method of item 2139 wherein the device is adapted for intrathecal delivering baclofen.
2143. The method of item 2139 wherein the device further comprises an intraspinal catheter. 2144. The method of item 2139 further comprising a second intrathecal drug delivery pump.
2145. The method of item 2139 wherein the device further comprises a catheter and an electrode.
2146. The method of any one of items 1896-2106 wherein the device is an implantable drug delivery pump for chemotherapy.
2147. The method of item 2146 wherein the device is adapted for delivering 2'-deoxy 5-fluorouridine.
2148. The method of item 2146 wherein the host has a solid tumor, and the device is adapted for infusing a chemotherapeutic agent to the solid tumor.
2149. The method of item 2146 wherein the host has a tumor, and the device is adapted for infusing a chemotherapeutic agent to the blood vessels that supply the tumor.
2150. The method of item 2146 wherein the host has a hepatic tumor, and the device is adapted for delivering a chemotherapeutic agent to the artery that provides blood supply to the liver of the host.
2151. The method of any one of items 1896-2106 wherein the device is a drug delivery pump for treating heart disease.
2152. The method of item 2151 wherein the device is an implantable cardiac electrode that delivers stimulation energy and dispenses drug adjacent to the stimulation site. The following examples are offered by way of illustration, and not by way of limitation.
EXAMPLES
EXAMPLE 1 PARYLENE COATING
A metallic portion of a housing of the device (e.g., MiniMed 2007 implantable insulin pump, Medtronic, Inc.) is washed by dipping it into HPLC grade isopropanol. A parylene primer layer (about 1 to 10 um) is deposited onto the cleaned device using a parylene coater (e.g., PDS 2010 LABCOATER 2 from Cookson Electronics) and di-p-xylylene (PARYLENE N) or dichloro-di-p- xylylene (PARYLENE D) (both available from Specialty Coating Systems, Indianapolis, IN) as the coating feed material.
EXAMPLE 2 JUGLONE COATING - PARTIAL COATING
Juglone solutions are prepared by dissolving Juglone (5 mg, 10 mg, 50 mg, 100 mg, 200 mg and 500 mg) in 5 ml HPLC grade THF. A coated portion of a parylene-coated device (as prepared in, e.g., Example 1) is dipped into a Juglone/THF solution. After a selected incubation time, the device is removed from the solution and dried in a forced air oven (5O0C). The device then is further dried in a vacuum oven overnight. The amount of Juglone used in each solution and the incubation time are varied such that the amount of Juglone coated onto the device is in the range of 0.06 μg/mm2 to 10 μg/mm2 (μg Jugione/mm2 of the device which is coated with Juglone after being placed in the THF/Juglone solution). The time during which the device is maintained in the Juglone/THF solution may be varied, where longer soak times generally provide for more Juglone to be adsorbed onto the device. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN- 5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 3
JUGLONE COATING - COMPLETE COATING
Juglone solutions are prepared by dissolving Juglone (5 mg, 10 mg, 50 mg, 100 mg, 200 mg and 500 mg) in 5 ml HPLC grade THF. An entire parylene coated device (coated as in, e.g., Example 1) is then dipped into the Juglone/THF solution. After a selected incubation time, the device is removed and dried in a forced air oven (5O0C). The device is then further dried in a vacuum oven overnight. The amount of Juglone used in each solution and the incubation time are varied such that the amount of Juglone coated onto the device is in the range of 0.06 μg/mm2 to 10 μg/mm2. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin. EXAMPLE 4
APPLICATION OF A PARYLENE OVERCOAT
A juglone coated device (prepared as in, e.g., Example 2 or 3) is placed in a parylene coater and an additional thin layer of parylene is deposited on the Juglone coated device using the procedure described in Example 1. The coating duration is selected to provide a parylene top-coat thickness that will cause the device to have a desired elution profile for the juglone.
EXAMPLE 5 APPLICATION OF AN ECHOGENIC COATING LAYER
DESMODUR (an isocyanate pre-polymer, Bayer AG) (25% w/v) is dissolved in a 50:50 mixture of dimethylsulfoxide and tetrahydrofuran. A Juglone/parylene overcoated device (prepared as in, e.g., Example 4) is then dipped into the pre-polymer solution. The device is removed from the solution after a selected incubation time, and the coating is then partially dried at room temperature for 3 to 5 minutes. The device is then immersed in a beaker of water (room temperature) for 3-5 minutes to cause the polymerization reaction to occur rapidly. An echogenic coating is formed.
EXAMPLE 6 JUGLONE/POLYMER COATING - PARTIAL COATING
Several 5% solutions of poly(ethylene-co-vinyl acetate) {EVA} (60% vinyl acetate) are prepared using THF as the solvent. Selected amounts of Juglone (0.01%, 0.05%, 0.1%, 0.5%, 1%, 5%, 10%, 20%, 30% (w/w drug to polymer) are added to the EVA solutions. The catheter portion of an implantable pump device or a portion thereof is dipped into a Juglone/EVA solution. The device is removed from the solution, and the coating is dried by placing the device in a forced air oven (400C) for 3 hours. The coated device is then further dried under vacuum for 24 hours. This dip coating process may be repeated to increase the amount of polymer/Juglone coated onto the device. In addition, higher Juglone concentrations in the polymer/THF/Juglone solution and/or a longer soak time may be used to increase the amount of polymer/Juglone that is coated onto the device. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 7 JUGLONE-HEPARIN COATING
Several 5% solutions of poly(ethylene-co-vinyl acetate) {EVA} (60% vinyl acetate) are prepared using THF as the solvent. Selected amounts (0.01 %, 0.05%, 0.1%, 0.5%, 1%, 5%, 10%, 20%, 30% (w/w drug to polymer)) of juglone and a solution of tridodecyl methyl ammonium chloride-heparin complex (PolySciences) are added to each of the EVA solutions. All, or a portion of, a catheter portion of the device is dipped into the juglone/EVA solution. After removing the device from the solution, the coating is dried by placing the device in a forced air oven (4O0C) for 3 hours. The coated device is then further dried under vacuum for 24 hours. The dip coating process may be repeated to increase the amount of polymer/heparin complex coated onto the device. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 8 JUGLONE - HEPARIN/HEPARIN COATING
An uncoated portion of a juglone-heparin coated device (prepared as in, e.g., Example 7) is dipped into a 5% EVA/THF solution containing a selected amount of a tridodecyl methyl ammonium chloride-heparin complex solution (PolySciences) (0.1%, 0.5%, 1%, 2.5%, 5%, 10% (v/v)). After removing the device from the solution, the coating is dried by placing the device in a forced air oven (4O0C) for 3 hours. The coated device is then further dried under vacuum for 24 hours. This provides a device with a Juglone/heparin coating on one or more portions of the device and a heparin coating on one or more other parts of the device. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin. EXAMPLE 9 JUGLONE/POLYMER COATING - PARTIAL COATING
Several 5% solutions of poly(styrene-co-isobutylene-styrene) (SIBS) are prepared using THF as the solvent. A selected amount of Juglone is added to each SIBS solution. One or more portions of the catheter portion of an implantable pump device are dipped into the juglone/SIBS solution. After removing the device from the solution, the coating is dried by placing the device in a forced air oven (4O0C) for 3 hours. The coated device is then further dried under vacuum for 24 hours. The dip coating process may be repeated to increase the amount of polymer/Juglone coated onto the device. In addition, higher juglone concentrations in the polymer/THF/juglone solution and/or a longer soak time may be used to increase the amount of polymer/Juglone that is coated onto the device. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatin.
EXAMPLE 10 JUGLONE/POLYMER COATING - ECHOGENIC OVERCOAT
A juglone-coated device prepared as described in Example 9 is dipped into a DESMODUR solution (50% w/v) (50:50 mixture of dimethylsulfoxide and tetrahydrofuran). The device is then removed and the coating is partially dried at room temperature for 3 to 5 minutes. The device is then immersed in a beaker of water (room temperature) for 3-5 minutes to cause the polymerization reaction to occur rapidly. An echogenic coating is thereby formed. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 11 POLYMER/ECHOGENIC COATING
A 5% solution of poly(styrene-co-isobutylene-styrene) (SIBS) is prepared using THF as the solvent. The catheter portion of an implantable pump device is dipped into the SIBS solution. After a selected incubation time, the device is removed from the solution, and the coating is dried by placing the device in a forced air oven (400C) for 3 hours. The coated device is then further dried under vacuum for 24 hours.
A coated device is dipped into a DESMODUR solution (50:50 mixture of dimethylsulfoxide and tetrahydrofuran). The device is then removed and the coating is then partially dried at room temperature for 3 to 5 minutes. The device is then immersed in a beaker of water (room temperature) for 3-5 minutes to cause the polymerization reaction to occur rapidly. The device is dried under vacuum for 24 hours at room temperature. All or a portion of the coated device is immersed into a solution of Juglone (5% w/v in methanol). The device is removed and dried at 400C for 1 hour and then under vacuum for 24 hours. The amount of juglone absorbed by the polymeric coating can be altered by changing the juglone concentration and the immersion time, as well as the solvent composition of the juglone solution. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 12 JUGLONE / SILOXANE COATING - PARTIAL COATING
The housing of an implantable pump device is coated with a silioxane layer by exposing the device to gaseous tetramethylcyclotetrasiloxane that is then polymerized by low energy plasma polymerization onto the device surface. The thickness of the siloxane layer can be increased by increasing the polymerization time. After polymerization, a portion of the coated device is then immersed into a juglone / THF solution (5% w/v) for a selected period of time to allow the Juglone to absorb into the siloxane coating. The device is then removed from the solution and is dried for 2 hours at 40°C in a forced air oven. The device is then further dried under vacuum at room temperature for 24 hours. The amount of juglone coated onto the device can be varied by altering the concentration of the juglone/THF solution and by altering the immersion time of the device in the juglone THF solution. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cervastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 13 SPRAY-COATED DEVICES
Several 2% solutions of poly(styrene-co-isobutylene-styrene) (SIBS) (50 ml) are prepared using THF as the solvent. A selected amount of juglone (0.01%, 0.05%, 0.1%, 0.5%, 1%, 2.5%, 5%, 10% and 20% (w/w with respect to the polymer)) is added to each solution. An implantable pump device is held with a pair of tweezers and is then spray coated with one of the juglone/polymer solutions using an airbrush. The device is then air-dried. The device is then held in a new location using the tweezers, and a second coat of a Juglone/polymer solution having the same concentration is applied to the device. The device is air-dried and is then dried under vacuum at room temperature overnight. The total amount of juglone coated onto the device can be altered by changing the juglone content in the solution, as well as by increasing the number of coatings that are applied. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin. EXAMPLE 14 DRUG COATED DEVICE-NON-DEGRADABLE
The catheter portion of an implantable pump device is attached to a rotating mandrel. A solution of Juglone (5% w/w) in a polyurethane (CHRONOFLEX 85A; CardioTech Biomaterials) / THF solution (2.5% w/v) is then sprayed onto all or a portion of the outer surface of the device. The solution is sprayed on at a rate that ensures that the device is not damaged or saturated with the sprayed solution. The device is allowed to air dry after which it is dried under vacuum for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP- 23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 15
DRUG COATED DEVICE- DEGRADABLE
The catheter portion of an implantable pump device is attached to a rotating mandrel. Juglone (5% w/w (juglone: PLGA)) in a PLGA/ethyl acetate solution (2.5% w/v) is then sprayed onto all or portion of the outer surface of the device. The solution is sprayed on at a rate that ensures that the device is not damaged or saturated with the sprayed solution. The device is allowed to air dry, after which it is dried under vacuum at room temperature for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 16 DRUG COATED DEVICE - DEGRADABLE OVERCOAT
A drug-coated catheter portion of an implantable pump device prepared as in Example 14 or Example 15 is attached to a rotating mandrel. A PLGA/ethyl acetate solution (2.5% w/v) is then sprayed onto all or a portion of the outer surface of the device, such that a coating is formed over the first drug- containing coating. The solution is sprayed on at a rate that ensures that the device is not damaged or saturated with the sprayed solution. The device is allowed to air dry, after which it is dried under vacuum at room temperature for 24 hours.
EXAMPLE 17 DRUG-LOADED MICROSPHERE FORMULATION
Juglone (10% w/w (juglone:PLGA)) is added to a solution of PLGA (50/50, Mw « 54,000) in DCM (5% w/v (PLGA:DCM)). The solution is vortexed and then poured into a stirred (overhead stirrer with a 3 bladed TEFLON coated stirrer) aqueous PVA solution (approx. 89% hydrolyzed, Mw « 13,000, 2% w/v). The solution is stirred for 6 hours after which the solution is centrifuged to sediment the microspheres. The microspheres are resuspended in water. The centrifugation process is repeated 4 times. The final microsphere solution is flash frozen in an acetone/dry-ice bath. The frozen solution is then freeze-dried to produce a fine powder. The size of the microspheres formed can be altered by changing the stirring speed and/or the PVA solution concentration. The freeze-dried powder can be resuspended in PBS or saline and can be used for direct injection, as an incubation fluid or as an irrigation fluid. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 18
DRUG COATED DEVICE (EXTERIOR COATING)
All or a portion of the catheter portion of an implantable pump device is dipped into a polyurethane (CHRONOFLEX 85A)/THF solution (2.5% w/v)). The coated device is allowed to air dry for 10 seconds. The device is then rolled in powdered Juglone that has been spread thinly on a piece of release liner to provide a device coated with between 0.1 to 10 mg of Juglone. The rolling process is done in such a manner that the Juglone powder predominantly adheres to the exterior side of the coated device. The device is air-dried for 1 hour followed by vacuum drying at room temperature for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, otepre no eta onate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 19
DRUG COATED DEVICE (EXTERIOR COATING) WITH A HEPARIN COATING
A drug-coated device prepared as in Example 18 is further coated with a heparin coating. A device prepared as in Example 18 is dipped into a solution of heparin-benzalkonium chloride complex (1.5% (w/v) in isopropanol, STS Biopolymers). The device is removed from the solution and air-dried for one hour, followed by vacuum drying for 24 hours. This process coats both the interior and exterior surfaces of the device with heparin.
EXAMPLE 20 PARTIAL DRUG COATING OF A DEVICE
The catheter portion of an implantable pump device is attached to a rotating mandrel. A mask system is set up so that only a portion of the device surface is exposed. A solution of Juglone (5% w/w) in a polyurethane (CHRONOFLEX 85A) / THF solution (2.5% w/v) is then sprayed onto the exposed portion of the device. The solution is sprayed on at a rate that ensures that the device is not damaged or saturated with the sprayed solution. The device is allowed to air dry, after which it is dried under vacuum at room temperature for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A1 brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol e a onate, pre n so one, puromyc n, - , c a r ne, mannose- - phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin..
EXAMPLE 21 DRUG - DEXAMETHASONE COATED DEVICE
The catheter portion of an implantable pump device is coated as in Example 20. The mask is then rearranged so that a previously masked portion of the device is exposed. The exposed portion of the device is then sprayed with a dexamethasone (10% w/w)/ polyurethane (CHRONOFLEX 85A)/THF solution (2.5% w/v)). The device is air dried, after which it is dried under vacuum at room temperature for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 22 DRUG - HEPARIN COATED DEVICE
The catheter portion of an implantable pump device is coated as in Example 20. The mask is then rearranged so that only a previously masked portion of the device is exposed. The exposed surface of the device is then sprayed with a heparin-benzalkonium chloride complex (1.5% (w/v) in isopropanol (STS Biopolymers). The sample is air-dried, after which it is dried under vacuum for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 23
DRUG-DEXAMETHAXONE COATED DEVICE
The catheter portion of an implantable pump device is attached to a rotating mandrel. A solution of Juglone (5% w/w) and dexamethazone (5% w/w) in a PLGA (50/50, Mw * 54,000) / ethyl acetate solution (2.5% w/v) is sprayed onto all or a portion of the device. The solution is sprayed on at a rate that ensures that the device is not damaged or saturated with the sprayed solution. The device is allowed to air dry, after which it is dried under vacuum at room temperature for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.. EXAMPLE 24
DRUG-DEXAMETHASONE COATED DEVICE (SEQUENTIAL COATING)
The catheter portion of an implantable pump device is attached to a rotating mandrel. A solution of juglone (5% w/w) in a PLGA (50/50, Mw « 54,000) / ethyl acetate solution (2.5% w/v) is sprayed onto the outer surface of the device. The solution is sprayed on at a rate that ensures that the device is not damaged or saturated with the sprayed solution. The device is allowed to air dry. A methanol solution of dexamethasone (2% w/v) is then sprayed onto the outer surface of the device (at a rate that ensures that the device is not damaged or saturated with the sprayed solution). The device is allowed to air dry, after which it is dried under vacuum at room temperature for 24 hours. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 25 DRUG-LOADING AN IMPLANTABLE GLUCOSE MONITOR - JUGLONE DIPPING
10 ml solutions of juglone are prepared by weighing 1 mg, 5 mg,
10 mg, 20 mg, 50 mg, 75 mg, 100 mg, 200 mg, and 500 mg juglone into separate 20 ml glass scintillation vials, respectively, and then adding HPLC grade methanol to each vial. The solutions are gently shaken on an orbital shaker for 1 hour at room temperature. The sensor tip of an implantable glucose sensor (DexCom, Inc.) is immersed to a depth of about 0.5 cm into the 0.1 mg/ml solution. After about 2 hours, the tip portion is removed from the solution and is allowed to air-dry for 6 hour. The electrode is further dried under vacuum for 24 hours. The process is repeated for all the prepared juglone solutions using a fresh sensor each time.
EXAMPLE 26
PREPARATION OF A DRUG-LOADED FILM FOR IMPLANTABLE GLUCOSE SENSORS -
NON-WOVEN MEMBRANES
353 ml dimethylacetamide (DMAC) is added to a 2 liter glass beaker. 660 g of a polyurethane solution (CHRONOFLEX AR, 25% solids in DMAC, CardioTech Biomaterials, Inc) is added to the solution. The solution is stirred for 15 min using an overhead stirrer unit (Cole Palmer) with a TEFLON coated paddle type stirrer blade. 62.5 g poly(vinylpyrrolidone) (PLASDONE K- 90D) is added to the solution. The solution is stirred for 6 hours until the polymers are all dissolved. Three sets of 5 x 15 g aliquots of the polymer solution are placed into 20 ml glass scintillation vials. To one set of the polymer solution, Juglone is added such that a Juglone to polymer ratio of 0.1%, 0.5%, 1 %, 10% and 20% is obtained. For the second set of the polymer solutions, rapamycin is added such that a rapamycin to polymer ratio of 0.1%, 0.5%, 1 %, 10% and 20% is obtained. For the third set of the polymer solutions, mythramycin is added such that a mythramycin to polymer ratio of 0.1%, 0.5%, 1 %, 10% and 20% is obtained. The solutions are tumbled for 3 hours at 20 rpm. A non-woven DACRON fiber filtration membrane is placed on a silicone coated PET release liner. A film is cast over the filter membrane from each of the polymer solutions using a casting knife (0.006"). The cast solutions are allowed to air dry for 1 hour at room temperature. The films are further dried at 5O0C for 3 hours after which they are dried under vacuum for 24 hours. Each film is cut to size and is mechanically secured to an implantable glucose sensing device (DexCom, Inc.) using an o-ring. PREPARATION OF A DRUG-LOADED FILM FOR IMPLANTABLE GLUCOSE SENSORS - POROUS MEMBRANES
353 ml dimethylacetamide (DMAC) is added to a 2L glass beaker. 660 g of a polyurethane solution (CHRONOFLEX AR, 25% solids in DMAC) is added to the solution. The solution is stirred for 15 min using an overhead stirrer unit (Cole Palmer) with a TEFLON coated paddle type stirrer blade. 62.5 g poly(vinylpyrrolidone) (PLASDONE K-90D) is added to the solution. The solution is stirred for 6 hours until the polymers are all dissolved. Three sets of 5 x 15 g aliquots of the polymer solution are placed into 20 ml glass scintillation vials. To one set of the polymer solution, Juglone is added such that a Juglone to polymer ratio of 0.1%, 0.5%, 1%, 10%, and 20% is obtained. For the second set of the polymer solutions, rapamycin is added such that a rapamycin to polymer ratio of 0.1%, 0.5%, 1 %, 10% and 20% is obtained. For the third set of the polymer solutions, mythramycin is added such that a mythramycin to polymer ratio of 0.1 %, 0.5%, 1%, 10%, and 20% is obtained. The solutions are tumbled for 3 hours at 20 rpm. A film of each of the polymer solutions is cast on a silicone coated PET release liner using a casting knife (0.012"). The cast solutions are allowed to air dry for 1 hour at room temperature. The films are further dried at 5O0C for 3 hours after which they are dried under vacuum for 24 hours. Each film is then pressed onto a porous silicone membrane (Seare Biomatrix Systems, Inc). Each film laminate is cut to size and is mechanically secured to an implantable glucose sensing device (DexCom, Inc.) using an o- ring.
EXAMPLE 28
DRUG-LOADING A MEMBRANE USED IN AN IMPLANTABLE GLUCOSE MONITOR - JUGLONE DIPPING
10 ml solutions of Juglone are prepared by weighing 1 mg, 5 mg, 10 mg, 20 mg, 50 mg, 75 mg, 100 mg, 200 mg, and 500 mg juglone into separate 20 ml glass scintillation vials, respectively, and then adding HPLC grade methanol to each vial. The solutions are gently shaken on an orbital shaker for 1 hour at room temperature. A CHRONOFLEX AR / PVP (Plasdone K-90D) (2.6:1 w/w) solution in DMAC is prepared as per Example 27. A non- woven DACRON fiber filtration membrane is placed on a silicone coated PET release liner. A film of the polymer solution is cast over the filter membrane using a casting knife. The cast solutions are allowed to air dry for 1 hour at room temperature. The films are further dried at 50°C for 3 hours after which they are dried under vacuum for 24 hours. A film is immersed in the 0.1 mg Juglone solution for 2 hours. The film is removed from the solution and is air dried for 2 hours at 450C. The film is then dried under vacuum for 24 hours. Each film is cut to size and is mechanically secured to an implantable glucose sensing device (DexCom, Inc.) using an o-ring. This process is repeated using all the prepared Juglone solutions. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 29 DRUG-LOADING A MEMBRANE USED IN AN IMPLANTABLE
GLUCOSE MONITOR - JUGLONE DIPPING
10 ml solutions of juglone are prepared by weighing 1 mg, 5 mg, 10 mg, 20 mg, 50 mg, 75 mg, 100 mg, 200 mg, and 500 mg Juglone into separate 20 ml glass scintillation vials, respectively, and then adding HPLC grade methanol to each vial. The solutions are gently shaken on an orbital shaker for 1 hour at room temperature. A CHRONOFLEX AR / PVP (PLASDONE K-90D) [2.6:1 w/w] film used in a implantable glucose monitoring device (DexCom, Inc.) is immersed in the 0.1 mg juglone solution for 2 hour. The film is removed from the solution and is air-dried for 2 hours at 45°C. The film is then dried under vacuum for 24 hours. Each film is then pressed onto a porous silicone membrane (Seare Biomatrix Systems, Inc). Each film laminate is cut to size and is mechanically secured to an implantable glucose sensing device (DexCom, Inc) using an o-ring. This process is repeated using all the prepared juglone solutions. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatin.
EXAMPLE 30 COATING OF A IMPLANTABLE GLUCOSE SENSOR
A polyurethane solution (CHRONOFLEX AL 85 A) is prepared by dissolving 20 g of the polyurethane in 400 ml tetrahydrofuran (THF). 15 ml aliquots of this solution are placed in 20 ml glass scintillation vials. 1 mg, 5 mg, 10 mg, 20 mg, 50 mg, 75 mg, 100 mg, and 200 mg juglone are then added to each of the vials, respectively. The solutions are tumbled for 3 hours at 20 rpm. An implantable glucose sensor device (DexCom, Inc.) is held in a clamp. The clamp is then attached to an overhead stirrer (Cole Palmer) and the stirring speed is set to 40 rpm. One of the juglone solutions is placed in a TLC spray device (Aldrich) that is attached to a nitrogen gas supply. The device is spray coated until a thin coating layer is obtained. The device is allowed to air dry for 5 hours. The device is removed from the clamp flipped 180 degrees and is again clamped. The coating process is then repeated. Each of the different juglone solutions is used to coat a separate device according to the coating process described above. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatin.
EXAMPLE 31 DRUG-LOADING THE CATHETER PORTION OF AN IMPLANTABLE PUMP -DIPPING
10 ml solutions of juglone are prepared by weighing 1 mg, 5 mg, 10 mg, 20 mg, 50 mg, 75 mg, 100 mg, 200 mg, and 500 mg juglone into separate 20 ml glass scintillation vials, respectively, and then adding HPLC grade methanol to each vial. The solutions are gently shaken on an orbital shaker for 1 hour at room temperature. The end segment of the catheter portion of an implantable pump (Medtronic) is immersed into the 0.1 mg/ml Juglone solution. After 2 hours the device is removed from the solution and is air dried for 24 hours at 37°C. Each of the different juglone solutions is used to coat a separate device according to the coating process described above. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatin.
EXAMPLE 32
COATING OF AN IMPLANTABLE PUMP
A polyurethane solution (CHRONOFLEX AL 85 A) is prepared by dissolving 20 g of polyurethane in 400 ml tetrahydrofuran (THF). 15 ml aliquots of this solution are placed in 20 ml glass scintillation vials. 1 mg, 5 mg, 10 mg, 20 mg, 50 mg, 75 mg, 100 mg, and 200 mg juglone are then added to each of the vials, respectively. The solutions are tumbled for 3 hours at 20 rpm. An implantable pump device (Medtronic, Inc) is held in a clamp. The clamp is then attached to an overhead stirrer (Cole Palmer) and the stirring speed is set to 40 rpm. One of the Juglone solutions is placed in a TLC spray device (Aldrich) that is attached to a nitrogen gas supply. The device is spray coated until a thin coating layer is obtained. The device is allowed to air dry for 5 hours. The device is removed from the clamp, flipped 180 degrees, and is again clamped. The coating process is then repeated. Each of the different juglone solutions is used to coat a separate device according to the coating process described above. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatin. EXAMPLE 33 DRUG-LOADING THE SENSOR PORTION OF A COCHLEAR IMPLANT -DIPPING
10 ml solutions of juglone are prepared by weighing 1 mg, 5 mg, 10 mg, 20 mg, 50 mg, 75 mg, 100 mg, 200 mg, and 500 mg juglone into separate 20 ml glass scintillation vials, respectively, and then adding HPLC grade methanol to each vial. The solutions are gently shaken on an orbital shaker for 1 hour at room temperature. The end segment of the sensor portion of a cochlear implant is immersed into the 0.1 mg/ml juglone solution. After 2 hours the device is removed from the solution and is air dried for 24 hours at 370C. Each of the different juglone solutions is used to coat a separate device according to the coating process described above. In additional examples, one of the following exemplary compounds may be used in lieu of juglone: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatin..
EXAMPLE 34
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS THERAPEUTIC AGENTS ON NITRIC OXIDE PRODUCTION BY MACROPHAGES
This example describes an assay for determining the effect of an agent on nitric oxide production by murine macrophages {(Chem. Ber. (1879) 12: 426; J. AOAC (1977) 60-594; Ann. Rev. Biochem. (1994) 63: 175). The murine macrophage cell line RAW 264.7 was trypsinized to remove cells from flasks, and the cells were plated in individual wells of a 6-well plate. Approximately 2 x 106 cells were plated in 2 ml of media containing 5% heat- inactivated fetal bovine serum (FBS). RAW 264.7 cells were incubated at 37°C for 1.5 hours to allow adherence to plastic. The agent was prepared in DMSO at a concentration of 10~2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10"2 M). Media was then removed and cells were incubated in 1 ng/ml of recombinant murine IFNγ and 5 ng/ml of LPS with or without mitoxantrone in fresh media containing 5% FBS. The agent was added to cells by directly adding agent DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Plates containing IFNy, LPS plus or minus the agent were incubated at 37°C for 24 hours.
At the end of the 24 hour period, supematants were collected from the cells and assayed for the production of nitrites. Each sample was tested in triplicate by aliquoting 50 μL of supernatant in a 96-well plate and adding 50 μL of Greiss Reagent A (0.5 g sulfanilamide, 1.5 ml H3PO4, 48.5 ml ddH2O) and 50 μL of Greiss Reagent B (0.05 g N-(1-naphthyl)-ethylenediamine, 1.5 ml H3PO4, 48.5 ml ddH2O). Optical density was read immediately using a microplate spectrophotometer at 562 nm absorbance. Absorbance over triplicate wells was averaged after subtracting background, and concentration values were obtained from the nitrite standard curve (1 μM to 2 mM). Inhibitory concentration of 50% (IC50) was determined by comparing average nitrite concentration to the positive control (cell stimulated with IFNy and LPS). An average of n=4 replicate experiments was used to determine IC50 values for the agent. IC50 values were obtained for the following compounds: herbimycin A (112 nM); geldanamycin (51 nM); 17-AAG (743 nM); 17-DMAG (50 nM); ruboxistaurin (2299 nM); actinomycin D (28 nM); CGP 74514A (700 nM); Radicicol from Humicola fuscoatra, (21 nM); loteprednol etabonate (221 nM); clobetasol propionate (99 nM); homoharringtonine (75 nM)); trichostatin A from Streptomyces sp. (310 nM); thapsigargin (33 nM); 3-BAABE (637 nM); jasplakinolide (153 nM). The following compounds showed no effect in this assay: simvastatin, tacrolimus, ribavarin, vinorelbine, pioglitazone, cladribine, pirfenidone, and mannose-6-phosphate. Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, juglone, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, brefeldin A, cerivastatin, dolastatin 15, prednisolone, puromycin, and 5- azacytidine.
EXAMPLE 35
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS ANTI-SCARRING AGENTS ON TNF-ALPHA PRODUCTION BY MACROPHAGES
This example describes an assay for determining the effect of an agent on TNF-α production by human macrophages (J. Immunol. (2000) 165: 411-418; J. Immunol. (2000) 164: 4804-4811 ; J. Immunol Meth. (2000) 235 (1- 2): 33-40). The human macrophage cell line, THP-1 was plated in a 12 well plate such that each well contained 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddHaO and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 xg and resuspended in 4 ml of human serum for a final concentration of 5 mg/ml. The resuspension was incubated in a 37°C water bath for 20 minutes to enable opsonization. Each agent was prepared in DMSO at a concentration of 10"2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10"2 M).
THP-1 cells were stimulated to produce TNFα by the addition of 1 mg/ml opsonized zymosan. The agent was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier as described above, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours.
After 24-hour stimulation, supernatants were collected to quantify TNFα production. TNFα concentrations in the supernatants were determined by ELISA using recombinant human TNFα to obtain a standard curve. A 96- well MaxiSorb plate was coated with 100 μl_ of anti-human TNFα Capture Antibody diluted in Coating Buffer (0.1 M sodium carbonate pH 9.5) overnight at 4°C. The dilution of Capture Antibody used was lot-specific and was determined empirically. Capture antibody was then aspirated and the plate washed 3 times with Wash Buffer (PBS1 0.05% TWEEN-20). Plates ere blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supernatants were diluted V8 and V^; (b) recombinant human TNFα was prepared at 500 pg/ml and serially diluted to yield a standard curve of 7.8 pg/ml to 500 pg/ml. Sample supernatants and standards were assayed in triplicate and were incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates were washed 5 times and incubated with 100 μL of Working Detector (biotinylated anti-human TNFα detection antibody + avidin-HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times, and 100 μL of Substrate Solution (tetramethylbenzidine, H2O2) was added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N H2SO4) was then added to the wells and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background was subtracted. TNFα concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC50) was determined by comparing average TNFα concentration to the positive control (THP- 1 cells stimulated with opsonized zymosan). An average of n=4 replicate experiments were used to determine IC50 values. IC50 values were obtained for the following compounds: herbimycin A (207 nM); geldanamycin (16 nM); 17-AAG (877 nM); 17-DMAG (112 nM); tacrolimus (0.5 nM); vinorelbine (244 nM); simvastatin (8876 nM); SB 203580 (171 nM); puromycin dihydrochloride from Streptomyces alboniger (751 nM); Radicicol from Humicola fuscoatra, ( 212 nM); homoharringtonine (32 nM); brefeldin A (53 nM); dolastatin 15 (28 nM). The following compounds showed no effect in this assay: ribavarin, pioglitazone, and mannose-6-phosphate. Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, clobetasol propionatetrichostatin A, thapsigargin, cerivastatin, jasplakinolide, pirfenidone, loteprednol etabonate, juglone, prednisolone, 3-BAABE, cladribine, 5-azacytidine, and Ly333531(ruboxistaurin).
EXAMPLE 36
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS THERAPEUTIC AGENTS
ON CELL PROLIFERATION
This example describes an assay for determining the effect of an agent on fibroblast cell proliferation ((In vitro toxicol. (1990) 3: 219; Biotech, Histochem. (1993) 68: 29; Anal. Biochem. (1993) 213: 426). Fibroblasts at 70- 90% confluency were trypsinized, replated at 600 cells/well in media in 96-well plates, and allowed to attach overnight. Each agent was prepared in DMSO at a concentration of 10'2 M and diluted 10-fold to give a range of stock concentrations (10"8 M to 10~2 M). Drug dilutions were diluted 1/1000 in media and added to cells to give a total volume of 200 μL/well. Each drug concentration was tested in triplicate wells. Plates containing fibroblasts and the agent were incubated at 37°C for 72 hours.
To terminate the assay, the media was removed by gentle aspiration. A 1/400 dilution of CYQUANT 400X GR dye indicator (Molecular Probes; Eugene, OR) was added to 1X Cell Lysis buffer, and 200 μL of the mixture was added to the wells of the plate. Plates were incubated at room temperature, protected from light for 3-5 minutes. Fluorescence was read in a fluorescence microplate reader at ~480 nm excitation wavelength and ~520 nm emission maxima. Inhibitory concentration of 50% (IC50) was determined by taking the average of triplicate wells and comparing average relative fluorescence units to the DMSO control. An average of n=4 replicate experiments was used to determine IC50 values. ICs0 values were obtained for the following compounds: herbimycin A (4.5 nM); geldanamycin (2.6 nM); 17- AAG (18 nM); 17-DMAG (6 nM); vinorelbine (4 nM); mboxostaurin (4626 nM); cladribine (184 nM); simvastatin (896 nM); actinomycin D (0.5 nM); juglone (314 nM); Bay 11-7085 (173 nM); puromycin dihydrochloride from Streptomyces alboniger (254 nM); decitabine (486 nM); penfluridol (878 nM); Radicicol from Humicola fuscoatra (26 nM); 5-azacytidine (806 nM); homoharringtonine (17 nM); trichostatin A from Streptomyces sp. (17 nM); brefeldin A (56 nM); thapsigargin (3 nM); AG-490 (623 nM); ammonium pyrrolidinedithiocarbamate (318 nM); dolastatin 15 (0.2 nM); cervistatin Na (33 nM); 3-BAABE (369 nM); jasplakinolide (3 nM); spermine tetrahydrochloride (283 nM). The following compounds showed no effect in this assay: tacrolimus, ribavarin, pioglitazone, pirfenidone, and mannose-6-phosphate. Other compounds that may be tested for IC5O values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, clobetasol propionate, loteprednol etabonate, and prednisolone.
EXAMPLE 37
EVALUATION OF JUGLONE CONTAINING MESH ON INTIMAL HYPERPLASIA
DEVELOPMENT IN A RAT BALLOON INJURY CAROTID ARTERY MODEL AS AN EXAMPLE
TO EVALUATE FIBROSIS INHIBITING AGENTS
A rat balloon injury carotid artery model is used to demonstrate the efficacy of a fibrosis-inhibiting agent (e.g., juglone) containing mesh system on the development of intimal hyperplasia fourteen days following placement. Control Group
Wistar rats weighing 400 - 500 g are anesthetized with 1.5% halothane in oxygen, and the left external carotid artery is exposed. An A 2 French FOGARTY balloon embolectomy catheter (Baxter, Irvine, CA) is advanced through an arteriotomy in the external carotid artery down the left common carotid artery to the aorta. The balloon is inflated with enough saline to generate slight resistance (approximately 0.02 ml) and it is withdrawn with a twisting motion to the carotid bifurcation. The balloon is then deflated and the procedure repeated twice more. This technique produces distension of the arterial wall and denudation of the endothelium. The external carotid artery is ligated after removal of the catheter. The right common carotid artery is not injured and is used as a control.
Local Perivascular Juαjone Treatment
Immediately after injury of the left common carotid artery, a 1 cm long distal segment of the artery is exposed and treated with a 1x1 cm juglone- containing mesh (345 ug juglone in a 50:50 PLG coating on a 10:90 PLG mesh - Example 61). The wound is then closed, and the animals are kept for 14 days.
Histology and immunohistochemistry At the time of sacrifice, the animals are euthanized with carbon dioxide and pressure perfused at 100 mmHg with 10% phosphate buffered formaldehyde for 15 minutes. Both carotid arteries are harvested and left overnight in fixative. The fixed arteries are processed and embedded in paraffin wax. Serial cross-sections are cut at 3 μm thickness every 2 mm within and outside the implant region of the injured left carotid artery and at corresponding levels in the control right carotid artery. Cross-sections are stained with Mayer's hematoxylin-and-eosin for cell count and with Movat's pentachrome stains for morphometry analysis and for extracellular matrix composition assessment. Other compounds that may be tested in this model include, e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), simvastatin geldanamycin, and 17-AAG,
EXAMPLE 38
EFFECT OF PACUTAXEL AND OTHER ANTI-MICROTUBULE AGENTS ON MATRIX METALLOPROTEINASE PRODUCTION
E. Materials and Methods
1. IL-1 stimulated AP- 1 transcriptional activity is inhibited by paclitaxel
Chondrocytes were transfected with constructs containing an AP- 1 driven CAT reporter gene, and stimulated with IL-1. IL-1 (50 ng/ml) was added and incubated for 24 hours in the absence and presence of paclitaxel at various concentrations. Paclitaxel treatment decreased CAT activity in a concentration dependent manner (mean ± SD) (see Figure 1B). The data noted with an asterisk (*) have significance compared with IL-1 -induced CAT activity according to a t-test, P<0.05. The results shown are representative of three independent experiments. 2. Effect of paclitaxel on IL-1 induced AP-1 DNA binding activity, AP- 1 DNA
Binding activity was assayed with a radiolabeled human AP-1 sequence probe and gel mobility shift assay. Extracts from chondrocytes untreated or treated with various amounts of paclitaxel (10'7 to 10~5 M) followed by IL-1 β (20 ng/ml) were incubated with excess probe on ice for 30 minutes, followed by non-denaturing gel electrophoresis. The "com" lane contains excess unlabeled AP-1 oligonucleotide (see Figure 1C). The results shown are representative of three independent experiments.
3. Effect of paclitaxel on IL-1 induced MMP-1 and MMP-3 mRNA expression
Cells were treated with paclitaxel at various concentrations (10"7 to 10"5 M) for 24 hours, then treated with IL-1β (20 ng/ml) for an additional 18 hours in the presence of paclitaxel. Total RNA was isolated, and the MMP-1 mRNA levels were determined by Northern blot analysis. The blots were subsequently stripped and reprobed with 32P-radiolabeled rat GAPDH cDNA, which was used as a housekeeping gene. The results shown are representative of four independent experiments (see Figure 1 D). Quantitation of collagenase-1 and stromelysin-expression mRNA levels were conducted. The MMP-1 and MMP-3 expression levels were normalized with GAPDH.
4. Effect of other anti-microtubules on collaqenase expression Primary chondrocyte cultures were freshly isolated from calf cartilage. The cells were plated at 2.5 x 106 per ml in 100 x 20 mm culture dishes and incubated in Ham's F12 medium containing 5% FBS overnight at 37 °C. The cells were starved in serum-free medium overnight and then treated with anti-microtubule agents at various concentrations for 6 hours. IL-1 (20 ng/ml) was then added to each plate and the plates incubated for an additional 18 hours. Total RNA was isolated by the acidified guanidine isothiocyanate method and subjected to electrophoresis on a denatured gel. Denatured RNA samples (15 μg) were analyzed by gel electrophoresis in a 1% denatured gel, transferred to a nylon membrane, and hydridized with the 32P-labeled collagenase cDNA probe. 32P-labeled glyceraldehyde phosphate dehydrase (GAPDH) cDNA was used as an internal standard to ensure roughly equal loading. The exposed films were scanned and quantitatively analyzed with IMAGEQUANT.
F. Results
1. Promoters on the family of matrix metalloproteinases
Figure 1A shows that all matrix metalloproteinases contained the transcriptional elements AP-1 and PEA-3 with the exception of gelatinase B. It has been well established that expression of matrix metalloproteinases such as collagenases and stromelysins are dependent on the activation of the transcription factor AP-1. Thus inhibitors of AP-1 may inhibit the expression of matrix metalloproteinases.
2. Effect of paclitaxel on AP-1 transcriptional activity
As demonstrated in Figure 1 B, IL-1 stimulated AP-1 transcriptional activity 5-fold. Pretreatment of transiently transfected chondrocytes with paclitaxel reduced IL-1-induced AP-1 reporter gene CAT activity. Thus, IL-1 induction of AP-1 activity was reduced in chondrocytes by paclitaxel in a concentration dependent manner (10"7 to 10'5 M). These data demonstrated that paclitaxel was a potent inhibitor of AP-1 activity in chondrocytes.
3. Effect of paclitaxel on AP-1 DNA binding activity
To confirm that paclitaxel inhibition of AP-1 activity was not due to nonspecific effects, the effect of paclitaxel on IL-1-induced AP-1 binding to oligonucleotides using chondrocyte nuclear lysates was examined. As shown in Figure 1C, IL-1 induced binding activity decreased in lysates from chondrocytes that had been pretreated with paclitaxel at concentrations from 10"7 to 10"5 M for 24 hours. Paclitaxel inhibition of AP- 1 transcriptional activity closely correlated with the decrease in AP-1 binding to DNA.
4. Effect of paclitaxel on collaqenase and stromelysin expression Since paclitaxel was a potent inhibitor of AP-1 activity, the effect of paclitaxel or IL-1 -induced collagenase and stromelysin expression, two important matrix metalloproteinases involved in inflammatory diseases, was examined. Briefly, as shown in Figure 1D, IL-1 induction increases collagenase and stromelysin mRNA levels in chondrocytes. Pretreatment of chondrocytes with paclitaxel for 24 hours significantly reduced the levels of collagenase and stromelysin mRNA. At 10"5 M paclitaxel, there was complete inhibition. The results show that paclitaxel completely inhibited the expression of two matrix metalloproteinases at concentrations similar to which it inhibits AP-1 activity.
5. Effect of other anti-microtubules on collagenase expression Figures 2A-H demonstrate that anti-microtubule agents inhibited collagenase expression. Expression of collagenase was stimulated by the addition of IL-1, which is a proinflammatory cytokine. Pre-incubation of chondrocytes with various anti-microtubule agents, specifically LY290181 , hexylene glycol, deuterium oxide, glycine ethyl ester, ethylene glycol bis- (succinimidylsuccinate), tubercidin, AIF3, and epothilone, all prevented IL-1- induced collagenase expression at concentrations as low as 1 x 10" M.
G. Discussion
Paclitaxel was capable of inhibiting collagenase and stromelysin expression in vitro at concentrations of 10"6 M. Since this inhibition may be explained by the inhibition of AP-1 activity, a required step in the induction of all matrix metalloproteinases with the exception of gelatinase B, it is expected that paclitaxel may inhibit other matrix metalloproteinases which are AP-1 dependent. The levels of these matrix metalloproteinases are elevated in all inflammatory diseases and play a principle role in matrix degradation, cellular migration and proliferation, and angiogenesis. Thus, paclitaxel inhibition of expression of matrix metalloproteinases such as collagenase and stromelysin can have a beneficial effect in inflammatory diseases. In addition to paclitaxel's inhibitory effect on collagenase expression, LY290181 , hexylene glycol, deuterium oxide, glycine ethyl ester, AIF3, tubercidin epothilone, and ethylene glycol bis-(succinimidylsuccinate), all prevented I L-1 -induced collagenase expression at concentrations as low as 1 x
7
10" M. Thus, anti-microtubule agents are capable of inhibiting the AP-1 pathway at varying concentrations. Other exemplary agents that may be tested in this assay include, juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531(ruboxistaurin), and simvastatin.
EXAMPLE 39 INHIBITION OF ANGIOGENESIS BY PACLITAXEL
Chick Chorioallantoic Membrane ("CAM") Assays
Fertilized, domestic chick embryos were incubated for 3 days prior to shell-less culturing. In this procedure, the egg contents were emptied by removing the shell located around the air space. The interior shell membrane was then severed and the opposite end of the shell was perforated to allow the contents of the egg to gently slide out from the blunted end. The egg contents were emptied into round-bottom sterilized glass bowls and covered with petri dish covers. These were then placed into an incubator at 90% relative humidity and 3% CO2 and incubated for 3 days.
Paclitaxel (Sigma, St. Louis, Ml) was mixed at concentrations of 0.25, 0.5, 1 , 5, 10, 30 μg per 10 μl aliquot of 0.5% aqueous methylcellulose. Since paclitaxel is insoluble in water, glass beads were used to produce fine particles. Ten microliter aliquots of this solution were dried on parafilm for 1 hour forming disks 2 mm in diameter. The dried disks containing paclitaxel were then carefully placed at the growing edge of each CAM at day 6 of incubation. Controls were obtained by placing paclitaxel-free methylcellulose disks on the CAMs over the same time course. After a 2 day exposure (day 8 of incubation) the vasculature was examined with the aid of a stereomicroscope. Liposyn II, a white opaque solution, was injected into the CAM to increase the visibility of the vascular details. The vasculature of unstained, living embryos was imaged using a Zeiss stereomicroscope that was interfaced with a video camera (Dage-MTI Inc., Michigan City, IN). These video signals were then displayed at 16Ox magnification and captured using an image analysis system (Vidas, Kontron; Etching, Germany). Image negatives were then made on a graphics recorder (Model 3000; Matrix Instruments, Orangeburg, NY). The membranes of the 8 day-old shell-less embryo were flooded with 2% glutaraldehyde in 0.1 M sodium cacodylate buffer; additional fixative was injected under the CAM. After 10 minutes in situ, the CAM was removed and placed into fresh fixative for 2 hours at room temperature. The tissue was then washed overnight in cacodylate buffer containing 6% sucrose. The areas of interest were postfixed in 1 % osmium tetroxide for 1.5 hours at 4°C. The tissues were then dehydrated in a graded series of ethanols, solvent exchanged with propylene oxide, and embedded in Spurr resin. Thin sections were cut with a diamond knife, placed on copper grids, stained, and examined in a Joel 1200EX electron microscope. Similarly, 0.5 mm sections were cut and stained with toluene blue for light microscopy. At day 11 of development, chick embryos were used for the corrosion casting technique. Mercox resin (Ted PeIIa, Inc., Redding, CA) was injected into the CAM vasculature using a 30-gauge hypodermic needle. The casting material consisted of 2.5 grams of Mercox CL-2B polymer and 0.05 grams of catalyst (55% benzoyl peroxide) having a 5 minute polymerization time. After injection, the plastic was allowed to sit in situ for an hour at room temperature and then overnight in an oven at 65°C. The CAM was then placed in 50% aqueous solution of sodium hydroxide to digest all organic components. The plastic casts were washed extensively in distilled water, air-dried, coated with gold/palladium, and viewed with the Philips 501 B scanning electron microscope.
Results of the assay were as follows. At day 6 of incubation, the embryo was centrally positioned to a radially expanding network of blood vessels; the CAM developed adjacent to the embryo. These growing vessels lie close to the surface and are readily visible making this system an idealized model for the study of angiogenesis. Living, unstained capillary networks of the CAM may be imaged noninvasively with a stereomicroscope.
Transverse sections through the CAM show an outer ectoderm consisting of a double cell layer, a broader mesodermal layer containing capillaries which lie subjacent to the ectoderm, adventitial cells, and an inner, single endodermal cell layer. At the electron microscopic level, the typical structural details of the CAM capillaries are demonstrated. Typically, these vessels lie in close association with the inner cell layer of ectoderm.
After 48 hours exposure to paclitaxel at concentrations of 0.25, 0.5, 1, 5, 10, or 30 μg, each CAM was examined under living conditions with a stereomicroscope equipped with a video/computer interface in order to evaluate the effects on angiogenesis. This imaging setup was used at a magnification of 16Ox which permitted the direct visualization of blood cells within the capillaries; thereby blood flow in areas of interest may be easily assessed and recorded. For this study, the inhibition of angiogenesis was defined as an area of the CAM (measuring 2-6 mm in diameter) lacking a capillary network and vascular blood flow. Throughout the experiments, avascular zones were assessed on a 4 point avascular gradient (Table 1). This scale represents the degree of overall inhibition with maximal inhibition represented as a 3 on the avascular gradient scale. Paclitaxel was very consistent and induced a maximal avascular zone (6 mm in diameter or a 3 on the avascular gradient scale) within 48 hours depending on its concentration.
Table 1 Avascular Gradient
0 - normal vascularity
1 — lacking some microvascular movement
2*- small avascular zone approximately 2 mm in diameter 3*— avascularity extending beyond the disk (6 mm in diameter)
* - indicates a positive antiangiogenesis response
The dose-dependent, experimental data of the effects of paclitaxel at different concentrations are shown in Table 1.
Table 1
Agent Delivery Vehicle Concentration Inhibition/n paclitaxel methylcellulose (10 ul) 0.25 ug 2/11 methylcellulose (10 ul) 0.5 ug 6/11 methylcellulose (10 ul) 1 ug 6/15 methylcellulose (10 ul) 5 ug 20/27 methylcellulose (10 ul) 10 ug 16/21 methylcellulose (10 ul) 30 ug 31/31
Typical paclitaxel-treated CAMs are also shown with the transparent methylcellulose disk centrally positioned over the avascular zone measuring 6 mm in diameter. At a slightly higher magnification, the periphery of such avascular zones is clearly evident; the surrounding functional vessels were often redirected away from the source of paclitaxel. Such angular redirecting of blood flow was never observed under normal conditions. Another feature of the effects of paclitaxel was the formation of blood islands within the avascular zone representing the aggregation of blood cells.
In summary, this study demonstrated that 48 hours after paclitaxel application to the CAM, angiogenesis was inhibited. The blood vessel inhibition formed an avascular zone which was represented by three transitional phases of paclitaxel's effect. The central, most affected area of the avascular zone contained disrupted capillaries with extravasated red blood cells; this indicated that intercellular junctions between endothelial cells were absent. The cells of the endoderm and ectoderm maintained their intercellular junctions and therefore these germ layers remained intact; however, they were slightly thickened. As the normal vascular area was approached, the blood vessels retained their junctional complexes and therefore also remained intact. At the periphery of the paclitaxel-treated zone, further blood vessel growth was inhibited which was evident by the typical redirecting or "elbowing" effect of the blood vessels. Exemplary compounds that may be tested in this model include: e.g., juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatingeldanamycin, and 17-AAG. EXAMPLE 40
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS AGENTS ON SMOOTH
MUSCLE CELL MIGRATION
This example describes an assay for determining the effect of an agent on human smooth muscle cell migration (Biotechniques (2000) 29: 81 ; J. Immunol. Methods (2001) 254: 85). Primary human smooth muscle cells were starved of serum in smooth muscle cell basal media containing insulin and human basic fibroblast growth factor (bFGF) for 16 hours prior to the assay. For the migration assay, cells were trypsinized to remove cells from flasks, washed with migration media, and diluted to a concentration of 2-2.5 X 105 cells/ml in migration media. Migration media consisted of phenol red free Dulbecco's Modified Eagle Medium (DMEM) containing 0.35% human serum albumin. A 100 μL volume of smooth muscle cells (approximately 20,000- 25,000 cells) was added to the top of a Boyden chamber assembly (Chemicon QCM CHEMOTAXIS 96-well migration plate). To the bottom wells, the chemotactic agent, recombinant human platelet derived growth factor (rhPDGF- BB), was added at a concentration of 10 ng/ml in a total volume of 150 μL. An agent (e.g., paclitaxel) was prepared in DMSO at a concentration of 10"2 M and serially diluted 10-fold to give a range of stock concentrations (10'8 M to 10"2 M). The agent was added to cells by directly adding the agent-DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to the cells in the top chamber. Plates were incubated for 4 hours to allow cell migration.
At the end of the 4 hour period, cells in the top chamber were discarded, and the smooth muscle cells attached to the underside of the filter were detached for 30 minutes at 37°C in Cell Detachment Solution (Chemicon). Dislodged cells were Iysed in lysis buffer containing the DNA binding CYQUANT GR dye and incubated at room temperature for 15 minutes. Fluorescence was read in a fluorescence microplate reader at ~480 nm excitation wavelength and ~520 nm emission maxima. Relative fluorescence units from triplicate wells were averaged after subtracting background fluorescence (control chamber without chemoattractant) and average number of cells migrating was obtained from a standard curve of smooth muscle cells serially diluted from 25,000 cells/well down to 98 cells/well. Inhibitory concentration of 50% (IC50) was determined by comparing the average number of cells migrating in the presence of the agent to the positive control (smooth muscle cell chemotaxis in response to rhPDGF-BB). IC50 values were obtained for the following compounds: paclitaxel (0.76 nM; see Figure 3); herbimycin A (738 nM); geldanamycin (86 nM); 17-AAG (429 nM); puromycin dihydrochloride from Streptomyces alboniger (324 nM); homoharringtonine (83 nM); cervistatin Na (90 nM). Simvastatin had no effect in this assay. Other compounds that may be tested in this assay include, e.g., juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, jasplakinolide, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, and Ly333531 (ruboxistaurin).
EXAMPLE 41 SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS COMPOUNDS ON IL-1 β
PRODUCTION BY MACROPHAGES
This example describes a screening assay for assessing the effect of an agent on IL-1β production by human macrophages. The assay protocol is based on the following references: J. Immunol. (2000) 165: 411- 418; J. Immunol. (2000) 164: 4804-4811 ; J. Immunol. Meth. (2000) 235 (1-2): 33-40). The human macrophage cell line, THP-1 was plated in a 12 well plate such that each well contained 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddH2O and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 g and resuspended in 4 ml of human serum for a final concentration of 5 mg/ml and incubated in a 37°C water bath for 20 minutes to enable opsonization. An agent was prepared in DMSO at a concentration of 10"2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10"2 M). THP-1 cells were stimulated to produce IL-1 β by the addition of 1 mg/ml opsonized zymosan. The agent was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours. After a 24 hour stimulation, supernatants were collected to quantify IL-1β production. IL-1β concentrations in the supernatants were determined by ELISA using recombinant human IL-1β to obtain a standard curve. A 96-well MaxiSorb plate was coated with 100 μl_ of anti-human IL-1β Capture Antibody diluted in Coating Buffer (0.1 M Sodium carbonate pH 9.5) overnight at 40C. The dilution of Capture Antibody used was lot-specific and was determined empirically. Capture antibody was then aspirated, and the plate was washed 3 times with Wash Buffer (PBS, 0.05% TWEEN-20). Plates were blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supernatants were diluted % and %; (b) recombinant human IL-1 β was prepared at 1000 pg/ml and serially diluted to yield a standard curve of 15.6 pg/ml to 1000 pg/ml. Sample supernatants and standards were assayed in triplicate and were incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates were washed 5 times and incubated with 100 μL of Working Detector (biotinylated anti-human IL-1β detection antibody + avidin-HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times, and 100 μL of Substrate Solution (tetramethylbenzidine, H2O2) was then added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N H2SO4) was then added to the wells and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background was subtracted. IL-1β concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC50) was determined by comparing average IL-1 β concentration to the positive control (THP-1 cells stimulated with opsonized zymosan). IC5O values were measured for the following compounds: herbimycin A (1027 nM); geldanamycin (267 nM); 17-AAG (253 nM); 17-DMAG (78 nM); vinorelbine (2359 nM); simvastatin (9572 nM), pirfenidone (80 nM); prednisolone 21- acetate (6.5 nM); NG-methyl-L-arginine acetate salt (<0.02 nM); tinidazole (96 nM); Radicicol from Humicola fuscoatra (369 nM); loteprednol etabonate (185 nM); homoharringtonine (7 nM); brefeldin A (42 nM). The following compounds showed no effect in this assay: mannose-6-phosphate, ribavarin, tacrolimus, pioglitazone, ruboxostaurin, and cladribine. Other compounds that may be tested for IC50 values in this assay include, e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, juglone, isotretinoin, clobetasol propionate, trichostatin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, hpuromycin, 3-BAABE, and 5-azacytidine.
EXAMPLE 42
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS COMPOUNDS ON IL-8
PRODUCTION BY MACROPHAGES
This example describes a screening assay for assessing the effect of an agent on IL-8 production by human macrophages. The assay protocol is based on the following references: J. Immunol. (2000) 165: 411- 418; J. Immunol. (2000) 164: 4804-4811 ; J. Immunol. Meth. (2000) 235 (1-2): 33-40. The human macrophage cell line, THP-1 was plated in a 12 well plate such that each well contained 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddH2O and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 g, resuspended in 4 ml of human serum for a final concentration of 5 mg/ml, and incubated in a 37°C water bath for 20 minutes to enable opsonization. The agent was prepared in DMSO at a concentration of 10"2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10"2 M).
THP-1 cells were stimulated to produce IL-8 by the addition of 1 mg/ml opsonized zymosan. An agent was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours.
After a 24 hour stimulation, supematants were collected to quantify IL-8 production. IL-8 concentrations in the supematants were determined by ELISA using recombinant human IL-8 to obtain a standard curve. A 96-well MAXISORB plate was coated with 100 μL of anti-human IL-8 Capture Antibody diluted in Coating Buffer (0.1 M sodium carbonate pH 9.5) overnight at 4°C. The dilution of Capture Antibody used was lot-specific and was determined empirically. Capture Antibody was then aspirated, and the plate was washed 3 times with Wash Buffer (PBS, 0.05% TWEEN-20). Plates were blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supematants were diluted 1Λoo and V1Ooo; (b) recombinant human IL-8 was prepared at 200 pg/ml and serially diluted to yield a standard curve of 3.1 pg/ml to 200 pg/ml. Sample supematants and standards were assayed in triplicate and were incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates were washed 5 times and incubated with 100 μL of Working Detector (biotinylated anti-human IL-8 detection antibody + avidin-HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times, and 100 μL of Substrate
Solution (tetramethylbenzidine, H2O2) was added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N H2SO4) iwass then added to the wells and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background was subtracted. IL-8 concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC50) was determined by comparing average IL-8 concentration to the positive control (THP-1 cells stimulated with opsonized zymosan). IC5O values were measured for the following compounds: herbimycin A (1431 nM); tacrolimus (188 nM); geldanamycin (19 nM); 17-AAG (209 nM); 17-DMAG (62 nM); mannose-6- phosphate (220 nM); simvastatin (9587 nM); isotretinoin (126 nM); puromycin dihydrochloride from Streptomyces alboniger (936 nM); NPC-15437 dihydrochloride (514 nM); Radicicol from Humicola fuscoatra (67 nM). The following compounds showed no effect in this assay: ribavarin, pirfenidone, vinorelbine, pioglitazone, ruboxostaurin, and cladribine. Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, juglone, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, loteprednol etabonate, prednisolone, 3-BAABE, and 5-azacytidine.
EXAMPLE 43
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS COMPOUNDS ON MCP- 1 PRODUCTION BY MACROPHAGES
This example describes a screening assay for assessing the effect of an agent on MCP-1 production by human macrophages. The assay protocol is based on the following references: J. Immunol. (2000) 165: 411- 418; J. Immunol. (2000) 164: 4804-4811 ; J. Immunol. Meth. (2000) 235 (1-2): 33-40. The human macrophage cell line, THP-1 was plated in a 12 well plate such that each well contained 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddh^O and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 g and resuspended in 4 ml of human serum for a final concentration of 5 mg/ml and incubated in a 37°C water bath for 20 minutes to enable opsonization. The agent was prepared in DMSO at a concentration of 10"2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10'2 M).
THP-1 cells were stimulated to produce MCP-1 by the addition of 1 mg/ml opsonized zymosan. An agent was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours.
After 24 hour stimulation, supernatants were collected to quantify MCP-1 production. MCP-1 concentrations in the supernatants were determined by ELISA using recombinant human MCP-1 to obtain a standard curve. A 96- well MaxiSorb plate was coated with 100 μl_ of anti-human MCP-1 Capture Antibody diluted in Coating Buffer (0.1 M sodium carbonate pH 9.5) overnight at 4°C. The dilution of Capture Antibody used was lot-specific and was determined empirically. Capture antibody as then aspirated and the plate washed 3 times with Wash Buffer (PBS, 0.05% TWEEN-20). Plates were blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supernatants were diluted 1/ioo and V-iooo; (b) recombinant human MCP-1 was prepared at 500 pg/ml and serially diluted to yield as standard curve of 7.8 pg/ml to 500 pg/ml. Sample supernatants and standards are assayed in triplicate and are incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates are washed 5 times and incubated with 100 μl_ of Working Detector (biotinylated anti-human MCP-1 detection antibody + avidin-HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times, and 100 μl_ of Substrate Solution (tetramethylbenzidine, H2O2) was added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N H2SO4) was then added to the wells, and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background was subtracted. MCP-1 concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC50) was determined by comparing average MCP-1 concentration to the positive control (THP-1 cells stimulated with opsonized zymosan). IC50 values were measured for the following compounds: herbimycin A (77 nM); geldanamycin (15 nM); 17- AAG (306 nM); 17-DMAG (59 nM); vinorelbine (38 nM); simvastatin (3031 nM); galardin (232 nM); idazoxan hydrochloride (15 nM); thioperamide maleate salt (63 nM); tirapazamine (554 nM); puromycin dihydrochloride from Streptomyces alboniger (161 nM); NPC-15437 dihydrochloride (402 nM); YM 976 (24 nM); Radicicol from Humicola fuscoatra (72 nM); risedronate (17 nM); aucubin (30 nM); homoharringtonine (8 nM); geniposidic acid (0.01 nM); geniposide (<0.01 nM); brefeldin A (33 nM); thapsigargin (4 nM); (-)-arctigenin (24 nM); dolastatin 15 (2 nM). The following compounds showed no effect in this assay: tacrolimus, mannose-6-phosphate, pioglitazone, ruboxistaurin, cladribine, ribavarin, and pirfenidone. Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, juglone, isotretinoin, clobetasol propionate, trichostatin A, cerivastatin, jasplakinolide, loteprednol etabonate, prednisolone, 3-BAABE, 5-azacytidine, and gefitinib. EXAMPLE 44
SCREENING ASSAY FOR ASSESSING THE EFFECT OF THERAPEUTIC AGENTS ON CELL
PROLIFERATION
This example describes a screening assay for assessing the effect of an agent on smooth muscle cell proliferation (In vitro toxicol. (1990) 3: 219; Biotech. Histochem. (1993) 68: 29; Anal. Biochem. (1993) 213: 426). This assay also may be used assess the effect of compounds on proliferation of fibroblasts and murine macrophage cell line RAW 264.7.
Smooth muscle cells at 70-90% confluency were trypsinized, replated at 600 cells/well in media in 96-well plates, and allowed to attach overnight. An agent {e.g., paclitaxel, geldanamycin, or herbimycin A) was prepared in DMSO at a concentration of 10"2 M and diluted 10-fold to give a range of stock concentrations (10~8 M to 10"2 M). Drug dilutions were diluted 1/1000 in media and added to cells to give a total volume of 200 μL/well. Each drug concentration was tested in triplicate wells. Plates containing cells and the agent were incubated at 37°C for 72 hours.
To terminate the assay, the media was removed by gentle aspiration. A 1/400 dilution of CYQUANT 400X GR dye indicator (Molecular Probes; Eugene, OR) was added to 1X Cell Lysis buffer, and 200 μL of the mixture was added to the wells of the plate. Plates were incubated at room temperature, protected from light for 3-5 minutes. Fluorescence was read in a fluorescence microplate reader at -480 nm excitation wavelength and ~520 nm emission maxima. Inhibitory concentration of 50% (IC50) was determined by taking the average of triplicate wells and comparing average relative fluorescence units to the DMSO control. An average of n=3 replicate experiments was used to determine IC50 values. ICs0 values were measured for the following compounds: herbimycin A (237 nM); geldanamycin (7.5 nM); 17- AAG (69 nM); 17-DMAG (17 nM); vinorelbine (15 nM); simvastatin (996 nM), ruboxistaurin (1825 nM), cladribine (137 nM); PD 98059 (<0.01 nM); actinomycin D (0.3 nM); juglone (902 nM); prednisolone 21-acetate (320 nM); puromycin dihydrochloride from Streptomyces alboniger (441 nM); dacarbazine (<0.01 nM); temozolomide (<0.01 nM); Radicicol from Humicola fuscoatra (34 nM); (-)-epicatechin gallate from green tea (428 nM); 5-azacytidine (542 nM); homoharringtonine (9 nM); trichostatin A from Streptomyces sp. (16 nM); NS- 398 (51 nM); betulinic acid (<0.01 nM); thapsigargin (5 nM); ammonium pyrrolidinedithiocarbamate (294 nM); dolastatin 15 (0.9 nM); cervistatin Na (12 nM); roxatidine acetate (<0.01 nM); benzoyl peroxide (<0.01 nM); 3-BAABE (403 nM); jasplakinolide (3 nM). The following compounds showed no effect in this assay: pioglitazone, mannose-6-phosphate, tacrolimus, ribavarin, and pirfenidone. Other compounds that may be tested for IC5O values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, clobetasol propionate, brefeldin A, and loteprednol etabonate.
EXAMPLE 45 PREPARATION OF DRUG-LOADED POLYURETHANE (PU) FILMS
Two g CHRONOFLEX - AL 85A (CT Biomaterials, Woburn, Mass.) Is added to a 20 ml glass scintillation vial. 10 ml THF are added together with a magnetic stirrer bar, and the vial is capped with the screw top lid. The mixture is stirred using a magnetic stirrer at a temperature of 50 0C until the polymer is dissolved. The solution is allowed to cool and the desired mass of the drug (10mg, 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, and 500 mg) is added to the solution. The solution is stirred to ensure proper mixing of the drug. Depending on the drug solubility in the THF, the drug may be dissolved in a suitable organic solvent prior to addition to the polymer solution. Solvents that can be used include DCM, acetone, dimethylacetamide, and ethanol. Depending on the drug, these mixtures may be solutions or may have the some or all of the drug in suspension. The mixture is then poured onto a sheet of release liner (rexam a10, grade 10393, silicone coated PET), and a casting knife (40 mil) is used to draw the mixture into a film. The film is covered with a sheet of glass (setup in such a manner that the glass is about 10mm from the wet film, that is, not touching the wet film) and is allowed to air dry for 10 min. The glass sheet is removed and the film is air dried in a hood for a further 60 min. The film is then placed in a forced air oven (50 0C) for 2 hours after which the film is placed in a vacuum oven for 24-48 hours. Once dried, the film is covered with another sheet of the release liner. This release liner laminated film is then cut into squares of approx. 8 mm x 8 mm. The films are then placed in a plastic pouch which is heat sealed closed. The films are then sterilized using either gamma sterilization or e-beam sterilization. Films containing the following drugs may be made using the described process: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, tacrolimus, juglone, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 46
PERIVASCULAR ADMINISTRATION OF THERAPEUTIC COMPOUNDS TO ASSESS
INHIBITION OF FIBROSIS
WISTAR rats weighing 250 - 300 g are anesthetized by the intramuscular injection of Innovar (0.33 ml/kg). Once sedated, they are then placed under Halothane anesthesia. After general anesthesia is established, fur over the neck region is shaved, the skin clamped and swabbed with betadine. A vertical incision is made over the left carotid artery and the external carotid artery exposed. Two ligatures are placed around the external carotid artery and a transverse arteriotomy is made. A number 2 French Fogarty balloon catheter is then introduced into the carotid artery and passed into the left common carotid artery and the balloon is inflated with saline. The catheter is passed up and down the carotid artery three times. The catheter is then removed and the ligature is tied off on the left external carotid artery.
A drug-loaded PU film (see Example 45) is then placed in a circumferential fashion around the common carotid artery. Five rats from each group are sacrificed at 14 days and the final five rats are sacrificed at 28 days. The rats are observed for weight loss or other signs of systemic illness. After 14 or 28 days, the animals are anesthetized and the left carotid artery is exposed in the manner of the initial experiment. The carotid artery is isolated, fixed at 10% buffered formaldehyde, and examined for histology. A statistically significant reduction in the degree of initimal hyperplasia, as measured by standard morphometric analysis, indicates a drug-induced reduction in fibrotic response. Compounds that may be tested in this model include, juglone, ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531(ruboxistaurin) and simvastatin.
EXAMPLE 47
IN VIVO EVALUATION OF SILK COATED PERIVASCULAR PU FILMS TO ASSESS THE ABILITY OF AN AGENT TO INDUCE SCARRING
A rat carotid artery model is described for determining whether a substance stimulates fibrosis. Wistar rats weighing 30Og to 40Og are anesthetized with halothane. The skin over the neck region is shaved and the skin is sterilized. A vertical incision is made over the trachea and the left carotid artery is exposed. A polyurethane film covered with silk strands or a control uncoated PU film is wrapped around a distal segment of the common carotid artery. The wound is closed and the animal is allowed to recover. After 28 days, the rats are sacrificed with carbon dioxide and pressure-perfused at 100 mmHg with 10% buffered formaldehyde. Both carotid arteries are harvested and processed for histology. Serial cross-sections can be cut every 2 mm in the treated left carotid artery and at corresponding levels in the untreated right carotid artery. Sections are stained with H&E and Movat's stains to evaluate tissue growth around the carotid artery. Area of perivascular granulation tissue is quantified by computer-assisted morphometric analysis. Area of the granulation tissue is significantly higher in the silk coated group than in the control uncoated group. See Figure 4.
EXAMPLE 48
IN VIVO EVALUATION OF PERIVASCULAR PU FILMS COATED WITH DIFFERENT SILK SUTURE MATERIAL TO ASSESS SCARRING
A rat carotid artery model is described for determining whether a substance stimulates fibrosis. Wistar rats weighing 30Og to 40Og are anesthetized with halothane. The skin over the neck region is shaved and the skin is sterilized. A vertical incision is made over the trachea and the left carotid artery is exposed. A polyurethane film covered with silk sutures from one of three different manufacturers (3-0 Silk - Black Braided (Davis & Geek), 3-0 SOFSILK (U.S. Surgical/ Davis & Geek), and 3-0 Silk -Black Braided (LIGAPAK) (Ethicon, Inc.) is wrapped around a distal segment of the common carotid artery. (The polyurethane film can also be coated with other agents to induce fibrosis.) The wound is closed and the animal is allowed to recover. After 28 days, the rats are sacrificed with carbon dioxide and pressure-perfused at 100 mmHg with 10% buffered formaldehyde. Both carotid arteries are harvested and processed for histology. Serial cross-sections are cut every 2 mm in the treated left carotid artery and at corresponding levels in the untreated right carotid artery. Sections are stained with H&E and Movat's stains to evaluate tissue growth around the carotid artery. Area of perivascular granulation tissue is quantified by computer-assisted morphometric analysis. Thickness of the granulation tissue is the same in the three groups showing that tissue proliferation around silk suture is independent of manufacturing processes. See Figure 5.
EXAMPLE 49
IN VIVO EVALUATION OF PERIVASCULAR SILK POWDER TO ASSESS THE CAPACITY OF AN AGENT TO INDUCE SCARRING
A rat carotid artery model is described for determining whether a substance stimulates fibrosis. Wistar rats weighing 30Og to 40Og were anesthetized with halothane. The skin over the neck region was shaved and the skin was sterilized. A vertical incision was made over the trachea and the left carotid artery was exposed. Silk powder was sprinkled on the exposed artery that was then wrapped with a PU film. Natural silk powder or purified silk powder (without contaminant proteins) was used in different groups of animals. Carotids wrapped with PU films only were used as a control group. The wound was closed and the animal was allowed to recover. After 28 days, the rats were sacrificed with carbon dioxide and pressure-perfused at 100 mmHg with 10% buffered formaldehyde. Both carotid arteries were harvested and processed for histology. Serial cross-sections can be cut every 2 mm in the treated left carotid artery and at corresponding levels in the untreated right carotid artery. Sections were stained with H&E and Movat's stains to evaluate tissue growth around the carotid artery. Area of tunica intima, tunica media, and perivascular granulation tissue was quantified by computer-assisted morphometric analysis. The natural silk caused a severe cellular inflammation consisting mainly of a neutrophil and lymphocyte infiltrate in a fibrin network without any extracellular matrix or blood vessels. In addition, the treated arteries were seriously damaged with hypocellular media, fragmented elastic laminae and thick intimal hyperplasia, lntimal hyperplasia contained many inflammatory cells and was occlusive in 2/6 cases. This severe immune response was likely triggered by antigenic proteins coating the silk protein in this formulation. On the other end, the regenerated silk powder triggered only a mild foreign body response surrounding the treated artery. This tissue response was characterized by inflammatory cells in extracellular matrix, giant cells and blood vessels. The treated artery was intact. These results show that removing the coating proteins from natural silk prevents the immune response and promotes benign tissue growth. Degradation of the regenerated silk powder was underway in some histology sections indicating that the tissue response can likely mature and heal over time. See Figure 6.
EXAMPLE 50
IN VIVO EVALUATION OF PERIVASCULAR TALCUM POWDER TO ASSESS THE CAPACITY OF AN AGENT TO INDUCE SCARRING
A rat carotid artery model is described for determining whether a substance stimulates fibrosis. Wistar rats weighing 30Og to 400g were anesthetized with halothane. The skin over the neck region was shaved and the skin was sterilized. A vertical incision was made over the trachea and the left carotid artery was exposed. Talcum powder was sprinkled on the exposed artery that was then wrapped with a PU film. Carotids wrapped with PU films only were used as a control group. The wound was closed and the animal was recovered. After 1 or 3 months, the rats were sacrificed with carbon dioxide and pressure-perfused at 100 mmHg with 10% buffered formaldehyde. Both carotid arteries were harvested and processed for histology. Serial cross- sections were cut every 2 mm in the treated left carotid artery and at corresponding levels in the untreated right carotid artery. Sections were stained with H&E and Movat's stains to evaluate tissue growth around the carotid artery. Thickness of tunica intima, tunica media, and perivascular granulation tissue was quantified by computer-assisted morphometric analysis. Histopathology results and morphometric analysis showed the same local response to talcum powder at 1 month and 3 months. A large tissue reaction trapped the talcum powder at the site of application around the blood vessel. This tissue was characterized by a large number of macrophages within a dense extracellular matrix with few neutrophiles, lymphocytes and blood vessels. The treated blood vessel appeared intact and unaffected by the treatment. Overall, this result showed that talcum powder induced a mild long- lasting fibrotic reaction that was subclinical in nature and did not harm any adjacent tissue. See Figure 7.
EXAMPLE 51 MIC DETERMINATION BY MICROTITRE BROTH DILUTION METHOD
A. MIC assay of various gram negative and positive bacteria
MIC assays were conducted essentially as described by Amsterdam, D. 1996, "Susceptibility testing of antimicrobials in liquid media", p.52-111 , in Loman, V., ed. Antibiotics in laboratory medicine, 4th ed. Williams and Wilkins, Baltimore, MD. Briefly, a variety of compounds were tested for antibacterial activity against isolates of P. aeruginosa, K. pneumoniae, E. coli, S. epidermidus and S. aureus in the MIC (minimum inhibitory concentration assay under aerobic conditions using 96 well polystyrene microtitre plates (Falcon 1177), and Mueller Hinton broth at 370C incubated for 24h. (MHB was used for most testing except C721 (S. pyogenes), which used Todd Hewitt broth, and Haemophilus influenzae, which used Haemophilus test medium (HTM)) Tests were conducted in triplicate. The results are provided below in Table 2.
TABLE 2: MINIMUM INHIBITORY CONCENTRATIONS OF THERAPEUTIC AGENTS
Figure imgf002341_0001
Activities are in Molar concentrations Wt = wild type N = No activity
B. MIC of antibiotic-resistant bacteria
Various concentrations of the following compounds, mitoxantrone, cisplatin, tubercidin, methotrexate, 5-fluorouracil, etoposide, 2-mercaptopurine, doxorubicin, 6-mercaptopurine, camptothecin, hydroxyurea and cytarabine were tested for antibacterial activity against clinical isolates of a methicillin resistant S. aureus and a vancomycin resistant pediocoocus clinical isolate in an MIC assay as described above. Compounds which showed inhibition of growth (MIC value of <1.0x 10-3) included: mitoxantrone (both strains), methotrexate (vancomycin resistant pediococcus), 5-fluorouracil (both strains), etoposide (both strains), and 2-mercaptopurine (vancomycin resistant pediococcus). EXAMPLE 52 PREPARATION OF RELEASE BUFFER
The release buffer is prepared by adding 8.22 g sodium chloride, 0.32 g sodium phosphate monobasic (monohydrate) and 2.60 g sodium phosphate dibasic (anhydrous) to a beaker. 1 L HPLC grade water is added and the solution is stirred until all the salts are dissolved. If required, the pH of the solution is adjusted to pH 7.4 ± 0.2 using either 0.1 N NaOH or 0.1 N phosphoric acid.
EXAMPLE 53 RELEASE STUDY TO DETERMINE RELEASE PROFILE OF THE THERAPEUTIC AGENT
FROM A COATED DEVICE
A sample of the therapeutic agent-loaded catheter is placed in a 15 ml culture tube. 15 ml release buffer (Example 52) is added to the culture tube. The tube is sealed with a TEFLON lined screw cap and is placed on a rotating wheel in a 370C oven. At various time points, the buffer is withdrawn from the culture tube and is replaced with fresh buffer. The withdrawn buffer is then analyzed for the amount of therapeutic agent contained in this buffer solution using HPLC. EXAMPLE 54
SCREENING ASSAY TO ASSESS THE EFFECT OF COMPOUNDS ON MATRIX METALLOPROTEASE 1 ACTIVITY
Cultures of primary human dermal fibroblasts were grown to confluence, re-seeded in 12-well plates (50,000 cells/well), and grown to 80- 90% confluency. Prior to growth factor stimulation, cells were starved under low serum conditions (0.5% fetal bovine serum) for 24 hours. Cells were then stimulated with 10 ng/mL of platelet-derived growth factor-BB (PDGF-BB) and 1 ng/mL of IL-1 β for 72 hours. Compounds to be tested for Matrix
Metalloprotease 1 (MMP-1) inhibitory activity were added concurrently with growth factors at concentrations between 10"11 M and 10'5 M. After 72 hours, tissue culture media containing MMP-1 was collected and stored at -2O0C. To isolate MMP-1 , an MMP-1 specific monoclonal antibody that binds both the pro- and mature forms of MMP-1 was coated onto black 96-well microplates, and tissue culture media was added. After washing away unbound material, the pro- form of MMP-1 was converted to the mature, active form by addition of the activation reagent, p-aminophenylmercuric acetate (APMA). To assay for MMP-1 activity, a MMP-1 peptide substrate (200 μl_ of a 1mM solution) was added to the wells following a wash in wash buffer, and the plates were then incubated at 37°C for 20 hours in the dark. In the presence of active MMP-1 , the peptide was cleaved and the product was detected by a fluorescent microplate reader. Upon exitation at ~320 nm, the cleaved product will emit a fluorescent signal at ~405nm, which is proportional to the amount of MMP-1 specific enzymatic activity in the sample. Relative fluorescence units from triplicate wells were averaged after subtracting background fluorescence. Fluoresence units were converted to activity units utilizing a standard curve generated with recombinant pro-MMP-1 serially diluted from 12.5 ng/mL to 0.39 ng/mL. Inhibitory concentration of 50% (IC50) was determined for test compounds by comparing the average MMP-1 activity obtained in presence of compound to the positive control (supematants from cells stimulated in the absence of compound). From one to, optimally, four replicate experiments were conducted to determine average IC50 values. The IC50 of selected drugs on fibroblast MMP-1 activity were determined as follows: geldanamycin (1.3 nM); 17-AAG (30 nM); 17-DMAG (8 nM); simvastatin (no inhibitory activity); 1 a,25-dihydroxyvitamin D3, vitamin D3 (2249 nM); SP600125 (1869 nM); halofuginone hydrobromide, HBr (15 nM); fascaplysin (1073 nM); puromycin dihydrochloride from Streptomyces alboniger (626 nM); celastrol, celastrus scandens, tripterin (405 nM); imatinib mesilate, STI571 , CGP 57148B (109 nM); CPKC 412A, midostaurin, N-benzoylstaurosporine, PKC412A (80 nM); LBM642 (1032 nM); Radicicol from Humicola fuscoatra (700 nM); farglitazar, GI262570X (2197 nM); SC12267 (902 nM); homoharringtonine (26 nM); prednisolone 21- acetate (86 nM). Other exemplary compounds that may be tested in this assay include, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN- 5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, juglone, isotretinoin, clobetasol propionate, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, tacrolimus, temsirolimus, loteprednol etabonate, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, and Ly333531 (ruboxistaurin).
EXAMPLE 55
PREPARATION OF DRUG-LOADED MICROSPHERES BY SPRAY DRYING
3.6 grams of methoxy poly(ethylene glycol 5000))-block-(poly (DL- lactide). (65:35 MePEGPDLLA weight ratio) is dissolved in 200 ml methylene chloride. 100 mg of a drug, cerivastatin, that is previously sieved through a 100 μm stainless steel sieve, is added and the resulting solution is spray dried. Inlet temperature 5O0C, outlet temperature < 39°C, aspirator 100%, flow rate 700 l/hr. The collected microspheres are dried under vacuum at room temperature overnight to produce uniform, spherical particles having size ranges of less than about 10 microns (typically about 0.5 to about 2 microns). This process for preparing drug-containing microspheres may also be used with ZD-6474, AP- 23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, juglone, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6- phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin. EXAMPLE 56 DRUG-LOADED MICROSPHERES (<10 MICRON) BY AN EMULSION PROCESS
100 ml of freshly prepared 10% polyvinyl alcohol (PVA) solution is added into a 600 ml beaker. The PVA solution is stirred at 2000 rpm for 30 minutes. Meanwhile, 80 mg cerivastatin is added to a solution of 800 mg PLGA (50/50, IV= 0.35, Durect Corporation Pelham, AL) in 20 ml dichloromethane. This mixture is stirred for 20 min. The resultant mixture is added dropwise to the PVA solution while stirring at 2000 rpm with a Fisher DYNA-MIX stirrer. After addition is complete, the solution is allowed to stir for an additional 45 minutes. The microsphere solution is transferred to several disposable graduated polypropylene conical centrifuge tubes, washed with deionized water, and centrifuged at 2600 rpm for 10 minutes. The aqueous layer is decanted and the washing, centrifuging and decanting is repeated 3 times. The combined, washed microspheres are freeze-dried and vacuum dried to remove any excess water. This process for preparing drug-containing microspheres may also be used with juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin) and simvastatin. EXAMPLE 57 GELLING FORMULATION (PREMIX) I
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (50mg) and PEG-SH4 (tetra functional poly (ethylene glycol) thiol) (50 mg) (Sunbio, Inc.) (referred to as "premix"). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. The solid contents of syringe 1 and the acidic solution of syringe 2 are mixed through a mixing connector by repeatedly transferring the contents from one syringe to the other. After complete mixing, the entire mixture is pushed into one of the syringes. The syringe containing the mixture then is attached to one inlet of an applicator (MICROMEDICS air assisted spray-applicator (Model SA-6105)). Syringe 3 containing the pH 9.7 solution is attached onto another inlet of the applicator. The formulation is applied to a surface as specified by the applicator manufacturer. The composition can be applied to the device (e.g., MiniMed 2007 implantable insulin pump, Medtronic, Inc.) (a) immediately prior to implantation or (b) following implantation of the device (e.g., MiniMed 2007 implantable insulin pump, Medtronic, Inc.) the drug-loaded gel can be injected around the device or (c) to the tissue of the implantations site immediately prior to implanting the device.
EXAMPLE 58 CERIVASTATIN IN PREMIX
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SH4 (50 mg), PEG-SG4 (50 mg), and cerivastatin (5 mg, sifted to less than 100 micron using a 100 micron stainless steel sieve). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.35 ml 0.24 M monobasic sodium phosphate and 0.4 M sodium carbonate (pH 10.0) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 57. This process for preparing drug- containing in-situ forming hydrogels may also be used with juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 59 CERIVASTATIN LOADED MICROSPHERES IN PREMIX
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (50 mg), PEG-SH4 (50 mg), and 2.7% cerivastatin loaded MePEG5000-PDLLA (65:35) microspheres prepared by spray drying (0.5 or 2 mg) (prepared using the procedure described in Example 55). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 57. This process for preparing drug-containing in situ forming hydrogel may be used with juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 60 CERVASTATIN-LOADED GEL
A mass of microspheres (as prepared in Example 56) equivalent to 10 mg cerivastatin is added to a 3 ml_ plastic syringe which has been capped. 2 mL hyaluronic acid gel (SYNVISC, Genzyme, Cambridge, MA) is added to the 3 mL syringe. The contents of the drug/hyaluronic acid syringe are mixed through a mixing connector connected to a second syringe by repeatedly transferring the contents from one syringe to the other. After complete mixing, the entire mixture is pushed into one of the syringes. The syringe is removed from the mixing connector. The drug-loaded gel can be applied to the device (e.g., MiniMed 2007 implantable insulin pump, Medtronic, Inc.) immediately prior to implantation or following implantation of the device (e.g., MiniMed 2007 implantable insulin pump, Medtronic, Inc.) the drug-loaded gel can be injected around the device. This process for preparing drug- containing gel may also be used with juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin) and simvastatin.
EXAMPLE 61 JUGLONE-LOADED DEGRADABLE MESH
A PLGA mesh (10/90 - VICRYL Mesh [Catalogue : VKMM], Ethicon (Somerville, NJ)) of dimensions 5 cm x 5 cm is dipped into a solution of isopropanol (HPLC grade). The mesh is then dried under vacuum. 3 g PLGA (50/50, IV=O.15, Durect Corporation, Pelham, AL) is dissolved in 15 ml ethyl acetate (HPLC Grade, Fisher) in a 20 mL glass scintillation vial. 10.3 mg juglone is added to the polymer solution and the resultant mixture is vortexed for 10 minutes. The mesh is held with a pair of tweezers and is dipped into this polymer drug solution. The excess solution is removed by touching the mesh to the neck of the vial. The mesh is air dried by holding the coated mesh in front of a small electrical fan for 2-3 minutes. The mesh is placed in a PTFE Petri dish and is further dried in a fume hood for 2 hours. The coated mesh is further dried under vacuum in a vacuum oven for 24 hours. The coated mesh is then placed between 2 pieces of release liner (REXAM A10, 6 cm x 6 cm) after which it is sealed in a heat sealable plastic pouch. The mesh is then sterilized by gamma radiation. The mesh can then be used to wrap around all or a portion of the device (e.g., MiniMed 2007 implantable insulin pump, Medtronic, Inc.) immediately prior to use. This process for preparing drug-containing mesh may also be used with ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3- BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, paclitaxel Ly333531 (ruboxistaurin) and simvastatin.
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except by the appended items.

Claims

CLAIMS We item the following:
1. A device comprising (a) a sensor and (b) an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2. The device of claim 1 wherein the sensor is selected from a blood or tissue glucose monitor, a pressure sensor or stress sensor, a cardiac sensor, a respiratory sensor, an auditory sensor, and an electrolyte sensor or metabolite sensor.
3. A device comprising (a) a pump and (b) an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4. The device of claim 3 wherein the pump is selected from an implantable insulin pump, an intrathecal drug delivery pump, an implantable drug delivery pump; an implantable drug delivery pump for chemotherapy, and a drug delivery pump for treating heart disease.
5. The device of any one of claims 1-4 wherein the anti- scarring agent is an antimicrobial compound.
6. The device of claim 5 wherein the antimicrobial compound is brefeldin A.
7. The device of any one of claims 1-4 wherein the anti- scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an anti-proliferative agent, a diuretic, an anti-coagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an antineoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an α-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AX0R12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride.
8. The device of any one of claims 1-4 wherein the anti- scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyi transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbBi and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF- 1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
9. The device of any one of claims 1-4 wherein the anti- scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an\ microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and antineoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
10. The device of any one of claims 1 -4 wherein the anti- scarring agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
11. The device of any one of claims 1-10 further comprising a coating wherein the coating comprises (a) the anti-scarring agent or (b) the anti- scarring agent and a polymer.
12. The device of claim 11 wherein the coating is disposed on a surface of the device.
13. The device of claim 11 wherein the coating directly contacts the device or indirectly contacts the device.
14. The device of claim 11 wherein the coating partially covers the device or completely covers the device.
15. The device of claim 11 wherein the coating is selected from a uniform coating, non-uniform coating, a discontinuous coating, and a patterned coating.
16. The device of any one of claims 1-10 further comprising a coating wherein the anti-scarring agent is present in the coating in an amount ranging between (a) about 0.0001% to about 1% by weight; (b) about 1% to about 10% by weight; (c) about 10% to about 25% by weight; or (d) about 25% to about 70% by weight.
17. The device of any one of claims 1-10 further comprising a polymer or further comprising a polymeric carrier.
18. The device of claim 17 wherein the polymeric carrier comprises a copolymer, a block copolymer, a random copolymer, a biodegradable polymer, a non-biodegradable polymer, a hydrophilic polymer, a hydrophobic polymer, a polymer having hydrophilic domains, or a polymer having hydrophobic domains.
19. The device of any one of claims 1-10 further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer, an elastomer, a hydrogel, a silicone polymer, a hydrocarbon polymer, a styrene-derived polymer, a butadiene polymer, a macromer, a poly-ethylene glycol) polymer, and an amorphous polymer.
20. The device of any one of claims 1-10 further comprising a second pharmaceutically active agent.
21. The device of any one of claims 1-10 further comprising at least one of an anti-inflammatory agent, an agent that inhibits infection, anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, cisplatin, an anti-thrombotic agent, a visualization agent, and an echogenic material.
22. The device of claim 21 wherein the visualization agent is a radiopaque material or a MRl responsive material.
23. The device of claim 21 wherein the visualization agent comprises at least one of gadolinium chelate, iron, magnesium, manganese, copper, chromium, an oxide compound, a dye, a pigment, or a colorant.
24. The device of any one of claims 1-10 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
25. The device of any one of claims 1-10 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
26. The device of any one of claims 1-10 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
27. The device of any one of claims 1-10 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate, an increasing rate, or a decreasing rate.
28. The device of any one of claims 1-10 wherein the device comprises (a) about 0.01 μg to about 10 μg of the anti-scarring agent; (b) about 10 μg to about 10 mg of the anti-scarring agent; (c) about 10 mg to about 250 mg of the anti-scarring agent; (d) about 250 mg to about 1000 mg of the anti- scarring agent; or (e) about 1000 mg to about 2500 mg of the anti-scarring agent.
29. The device of any one of claims 1-10 wherein the device comprises (a) about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (c) about 10 μg to about 250 μg of the anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (d) about 250 μg to about 1000 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (e) about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
30. The device of any one of claims 1-10 wherein the device comprises (a) about 0.01 μg to about 100 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 0.01 μg to about 200 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (c) about 0.01 μg to about 500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
31. The device of any one of claims 1 , 2, and 5-10 wherein the agent or the composition is affixed to the sensor.
32. The device of any one of claims 1 , 2, and 5-10 wherein the agent or the composition is covaiently or non-covalently attached to the sensor.
33. The device of any one of claims 1-10 further comprising a coating that absorbs the agent or the composition.
34. The device of any one of claims 1 , 2, and 5-10 wherein (a) the sensor is interweaved with a thread composed of, or coated with, the agent or the composition; (b) the sensor is covered with a sleeve that contains the agent or the composition; or (c) the sensor is covered with a mesh that contains the agent or the composition.
35. The device of any one of claims 1-10 further comprising a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are the same or the first composition and the second composition are different.
36. The device of any one of claims 1-10 wherein the anti- scarring agent is located within pores or holes of the device or within a channel, lumen or divet of the device.
37. A method for inhibiting scarring comprising placing (a) a sensor and (b) an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
38. The method of claim 37 wherein the sensor is selected from a blood or tissue glucose monitor, a pressure sensor or stress sensor, a cardiac sensor, a respiratory sensor, an auditory sensor, and an electrolyte sensor or metabolite sensor.
39. A method for inhibiting scarring comprising placing (a) a pump and (b) an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
40. The method of claim 39 wherein the pump is selected from an implantable insulin pump, an intrathecal drug delivery pump, an implantable drug delivery pump; an implantable drug delivery pump for chemotherapy, and a drug delivery pump for treating heart disease.
41. The method of any one of claims 37-40 wherein the anti- scarring agent is an antimicrobial compound.
42. The method of any one of claims 37-40 wherein the antimicrobial compound is brefeldin A.
43. The method of any one of claims 37-40 wherein the anti- scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an antiproliferative agent, a diuretic, an anti-coagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an α-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride.
44. The method of any one of claims 37-40 wherein the anti- scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
45. The method of any one of claims 37-40 wherein the anti- scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an\ microtubuie inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and antineoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
46. The method of any one of claims 37-40 wherein the anti- scarring agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
47. The method of any one of claims 37-44 wherein the composition comprises a polymer or a polymeric carrier.
48. The method of any one of claims 37-44 wherein the composition comprises a polymer, and wherein the polymer is, or comprises, a copolymer, a block copolymer, a random copolymer, a biodegradable polymer, a non-biodegradabte polymer, a hydrophilic polymer, a hydrophobic polymer, a polymer having hydrophilic domains, or a polymer having hydrophobic domains, a non-conductive polymer, an elastomer, a hydrogel, a silicone polymer, a hydrocarbon polymer, a styrene-derived polymer, a butadiene polymer, a macromer, a poly-ethylene glycol) polymer, and an amorphous polymer.
49. The method of any one of claims 37-44 wherein the composition further comprises a second pharmaceutically active agent.
50. The method of any one of claims 37-44 wherein the composition further comprises at least one of an anti-inflammatory agent, an agent that inhibits infection, anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, cisplatin, an antithrombotic agent, a visualization agent, and an echogenic material.
51. The method of claim 50 wherein the visualization agent is a radiopaque material or a MRI responsive material.
52. The method of claim 50 wherein the visualization agent comprises at least one of gadolinium chelate, iron, magnesium, manganese, copper, chromium, an oxide compound, a dye, a pigment, or a colorant.
53. The method of any one of claims 37-44 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days or the agent is released in effective concentrations from the composition comprising the agent by erosion over a period ranging from the time of administration to about 90 days.
54. The method of any one of claims 37-44 wherein the composition further comprises an inflammatory cytokine or an agent that stimulates cell proliferation.
55. The method of any one of claims 37-44 wherein the composition is in the form of a gel, paste, or spray.
56. The method of any one of claims 37-38 and 41-44 wherein the sensor is partially constructed with the agent or the composition or wherein the sensor is impregnated with the agent or the composition.
57. The method of any one of claims 37-38 and 41-44 wherein the agent or the composition forms a coating, and the coating indirectly contacts the sensor or the coating directly contacts the sensor.
58. The method of any one of claims 37-38 and 41-44 wherein the agent or the composition forms a coating, wherein the coating partially covers the device or completely covers the device.
59. The method of any one of claims 37-38 and 41-44 wherein the agent or the composition is located within pores or holes of the sensor or wherein the agent or composition is located within a channel, lumen, or divet of the sensor.
60. The method of any one of claims 37-38 and 41-44 wherein the sensor further comprises a coating wherein the coating is a uniform coating, non-uniform coating, a discontinuous coating, and a patterned coating.
61. The method of any one of claims 37-38 and 41-44 wherein the sensor further comprises a coating, and the anti-scarring agent is present in the coating in an amount ranging between (a) about 0.0001 % to about 1% by weight; (b) about 1% to about 10% by weight; (c) about 10% to about 25% by weight; or (d) about 25% to about 70% by weight.
62. The method of any one of claims 37-38 and 41-44 wherein the agent is delivered from the sensor, wherein the agent is released into tissue in the vicinity of the device after deployment of the device.
63. The method of any one of claims 37-38 and 41-44 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
64. The method of any one of claims 37-38 and 41-44 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate, an increasing rate, or a decreasing rate.
65. The method of any one of claims 37-38 and 41-44 wherein the agent is delivered from the sensor, wherein the sensosr comprises (a) about 0.01 μg to about 10 μg of the anti-scarring agent; (b) about 10 μg to about 10 mg of the anti-scarring agent; (c) about 10 mg to about 250 mg of the anti-scarring agent; (d) about 250 mg to about 1000 mg of the anti-scarring agent; or (e) about 1000 mg to about 2500 mg of the anti-scarring agent.
66. The method of any one of claims 37-38 and 41-44 wherein the agent is delivered from the sensor, wherein the sensosr comprises (a) about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied; (c) about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (d) about 250 μg to about 1000 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (e) about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
67. The method of any one of claims 37-38 and 41-44 wherein the agent is delivered from the sensor, wherein the sensor comprises (a) about 0.01 μg to about 100 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 0.01 μg to about 200 μg of the anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied; or (c) about 0.01 μg to about 500 μg of the anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
68. The method of any one of claims 37-38 and 41-44 wherein wherein the agent or the composition is affixed to the sensor.
69. The method of any one of claims 37-38 and 41-44 wherein wherein the agent or the composition is covalently or non-covalently attached to the sensor.
70. The method of any one of claims 37-38 and 41-44 wherein the sensor further comprises a coating that absorbs the agent or the composition.
71. The method of any one of claims 37-38 and 41-44 wherein (a) the sensor is interweaved with a thread composed of, or coated with, the agent or the composition; (b) the sensor is covered with a sleeve that contains the agent or the composition; or (c) the sensor is covered with a mesh that contains the agent or the composition.
72. The method of any one of claims 37-38 and 41-44 wherein the sensor comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are the same or the first composition and the second composition are different.
73. The method of any one of claims 37-38 and 41-44 wherein the sensor is linked to a pump.
74. The method of any one of claims 37-38 and 41-44 wherein the agent or the composition is applied (a) to the sensor surface prior to the placing of the sensor into the host; (b) to the sensor surface during to the placing of the sensor into the host; or (c) to the sensor surface immediately after placing of the sensor into the host.
75. The method of any one of claims 37-38 and 41-44 wherein the agent or the composition is applied (a) to the surface of the tissue in the host surrounding the sensor prior to the placing of the sensor into the host; (b) to the surface of the tissue in the host surrounding the sensor during the placing of the sensor into the host; or (c) to the surface of the tissue in the host surrounding the sensor immediately after the placing of the sensor into the host.
76. The method of any one of claims 37-38 and 41-44 wherein the agent or the composition is topically applied into the anatomical space where the sensor is placed or is percutaneously injected into the tissue in the host surrounding the sensor.
77. A method for making a device comprising combining a sensor (a) a sensor and (b) an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
78. The method of claim 77 wherein the sensor is selected from a blood or tissue glucose monitor, a pressure sensor or stress sensor, a cardiac sensor, a respiratory sensor, an auditory sensor, and an electrolyte sensor or metabolite sensor.
79. A method for making a device comprising (a) a pump and (b) an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
80. The method of claim 79 wherein the pump is selected from an implantable insulin pump, an intrathecal drug delivery pump, an implantable drug delivery pump; an implantable drug delivery pump for chemotherapy, and a drug delivery pump for treating heart disease.
81. The method of any one of claims 77-80 wherein the anti- scarring agent is an antimicrobial compound.
82. The method of any one of claims 77-80 wherein the antimicrobial compound is brefeldin A.
83. The method of any one of claims 77-80 wherein the anti- scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an anti-proliferative agent, a diuretic, an anti-coagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an antineoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an anti- thrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an α-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno- modufator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride.
84. The method of any one of claims 77-80 wherein the anti- scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
85. The method of any one of claims 77-80 wherein the anti- scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an\ microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and antineoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
86. The method of any one of claims 77-80 wherein the anti- scarring agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsjgargin, dolastatin 15, cerivastatin, jasplakinofide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribiπe, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
87. The method of any one of claims 77-86 wherein the composition comprises a polymer or a polymeric carrier.
88. The method of any one of claims 77-86 wherein the device comprises a coating wherein the coating comprises (a) the anti-scarring agent or (b) the anti-scarring agent and a polymer.
89. The method of any one of claims 77-86 wherein the device comprises a coating that comprises the agent and wherein the coating is disposed on a surface of the device.
90. The method of any one of claims 77-86 wherein the device comprises a coating that comprises the agent and wherein the coating directly contacts the device or indirectly contacts the device.
91. The method of any one of claims 77-86 wherein the device comprises a coating that comprises the agent and the coating partially covers the device or completely covers the device.
92. The method of any one of claims 77-86 wherein the device comprises a coating that is a uniform coating, non-uniform coating, a discontinuous coating, or a patterned coating.
93. The method of any one of claims 77-86 wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between (a) about 0.0001% to about 1% by weight; (b) about 1% to about 10% by weight; (c) about 10% to about 25% by weight; or (d) about 25% to about 70% by weight.
94. The method of any one of claims 77-86 wherein the device has a coating, and wherein the coating further comprises a polymer.
95. The method of any one of claims 77-86 wherein the device comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are the same or the first composition and the second composition are different.
96. The method of any one of claims 77-86 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer, a block copolymer, a random copolymer, a biodegradable polymer, a non-biodegradable polymer, a hydrophilic polymer, a hydrophobic polymer, a polymer having hydrophilic domains, or a polymer having hydrophobic domains, a non-conductive polymer, an elastomer, a hydrogel, a silicone polymer, a hydrocarbon polymer, a styrene-derived polymer, a butadiene polymer, a macromer, a poly-ethylene glycol) polymer, and an amorphous polymer.
97. The method of any one of claims 77-86 wherein the device further comprises a second pharmaceutically active agent.
98. The method of any one of claims 77-86 wherein the device further comprises at least one of an anti-inflammatory agent, an agent that inhibits infection, anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, cisplatin, an antithrombotic agent, a visualization agent, and an echogenic material.
99. The method of claim 98 wherein the visualization agent is a radiopaque material or a MRl responsive material.
100. The method of claim 98 wherein the visualization agent comprises at least one of gadolinium chelate, iron, magnesium, manganese, copper, chromium, an oxide compound, a dye, a pigment, or a colorant.
101. The method of any one of claims 77-86 wherein the anti- scarring agent is located within pores or holes of the device or within a channel, lumen or divet of the device.
102. The method of any one of claims 77-86 wherein the anti- scarring agent is released locally to tissue proximate to the device.
103. The method of any one of claims 77-86 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
104. The method of any one of claims 77-86 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
105. The method of any one of claims 77-86 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate, an increasing rate, or a decreasing rate.
106. The method of any one of claims 77-86 wherein the device comprises (a) about 0.01 μg to about 10 μg of the anti-scarring agent; (b) about 10 μg to about 10 mg of the anti-scarring agent; (c) about 10 mg to about 250 mg of the anti-scarring agent; (d) about 250 mg to about 1000 mg of the anti-scarring agent; or (e) about 1000 mg to about 2500 mg of the anti- scarring agent.
107. The method of any one of claims 77-86 wherein the device comprises (a) about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (c) about 10 μg to about 250 μg of the anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (d) about 250 μg to about 1000 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (e) about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
108. The method of any one of claims 77-86 wherein the device comprises (a) about 0.01 μg to about 100 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 0.01 μg to about 200 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (c) about 0.01 μg to about 500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
109. The method of any one of claims 77-78 and 81-86 wherein the combining is performed by directly affixing the agent or the composition to the sensor.
110. The method of any one of claims 77-78 and 81-86 wherein the combining is performed by (a) spraying the agent or the composition onto the sensor; (b) electrospraying the agent or the composition onto the sensor; (c) dipping the sensor into a solution comprising the composition onto the sensor; (d) covalently attaching the agent or the composition to the sensor; (e) coating the sensor with a substance that contains the agent or the composition; (f) coating the sensor with a substance that absorbs the agent; (g) interweaving a thread composed of, or coated with, the agent or the composition; (h) completely covering the sensor with a sleeve that contains the agent or the composition; (i) covering a portion of the sensor with a sleeve that contains the agent or the composition; (j) completely covering the sensor with a cover that contains the agent or the composition; (k) covering a portion of the sensor with a cover that contains the agent or the composition; (I) completely covering the sensor with an electrospun fabric that contains the agent or the composition; (m) covering a portion of the sensor with an eiectrospun fabric that contains the agent or the composition; (n) completely covering the sensor with a mesh that contains the agent or the composition; (o) covering a portion of the sensor with a mesh that contains the agent or the composition; (p) constructing a portion of the sensor with the agent or the composition; (q) impregnating the sensor with the agent or the composition; (r) constructing a portion of the sensor from a degradable polymer that releases the agent; (s) clipping the sensor into a solution that comprise the agent and an inert solvent for the sensor; (t) dipping the sensor into a solution that comprises the agent and a solvent that will swell the sensor; (u) dipping the sensor into a solution that comprises the agent and a solvent that will dissolve the sensor; (v) dipping the sensor into a solution that comprises the agent, a polymer and an inert solvent for the sensor; (w) dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will swell the sensor; (x) dipping the sensor into a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor; (y) spraying the sensor with a solution that comprises the agent and an inert solvent for the sensor; (z) spraying the sensor with a solution that comprises the agent and a solvent that will swell the sensor; (aa) spraying the sensor with a solution that comprises the agent and a solvent that will dissolve the sensor; (bb) spraying the sensor with a solution that comprises the agent, a polymer and an inert solvent for the sensor; (cc) spraying the sensor with a solution that comprises the agent, a polymer and a solvent that will swell the sensor; or (dd) spraying the sensor with a solution that comprises the agent, a polymer and a solvent that will dissolve the sensor.
111. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, U) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a po)ymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a sensor.
112. The method of claim 111 wherein the sensor is selected from a blood or tissue glucose monitor, a pressure sensor or stress sensor, a cardiac sensor, a respiratory sensor, an auditory sensor, and an electrolyte sensor or metabolite sensor.
113. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a pump.
114. The method of claim 113 wherein the pump is selected from an implantable insulin pump, an intrathecal drug delivery pump, an implantable drug delivery pump; an implantable drug delivery pump for chemotherapy, and a drug delivery pump for treating heart disease.
115. The method of any one of claims 111-114 wherein the anti- fibrotic agent is an antimicrobial compound.
116. The method of any one of claims 111-114 wherein the antimicrobial compound is brefeldin A.
117. The method of any one of claims 111-114 wherein the anti- fibrotic agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an antiproliferative agent, a diuretic, an anti-coagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an α-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno- modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride.
118. The method of any one of claims 111-114 wherein the anti- fibrotic agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
119. The method of any one of claims 111-114 wherein the anti- fibrotic agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an\ microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and antineoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
120. The method of any one of claims 111-114 wherein the anti-fibrotic agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, s/inoreibine, 17-DMAG, tacrolimus, loteprednol etabonate, jugione, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
121. The method for implanting a medical device according any one of claims 111-120 comprising (a) infiltrating a tissue of a host where the medical device is to be, or has been implanted with (i) an anti-fibrotic agent; (ii) an anti-infective agent; (iii) a polymer; (iv); a composition comprising an anti- fibrotic agent and a polymer; (v) an anti-infective agent and a polymer; (vi) a composition comprising an anti-fibrotic agent, an anti-infective agent, and a polymer, and (b) implanting the medical device into a host.
122. The method for implanting a medical device according any one of claims 111 -120 wherein the anti-infective agent is selected from an anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, and cisplatin.
123. The method for implanting a medical device according any one of claims 111-120 wherein the composition comprises an antithrombotic agent.
124. The method for implanting a medical device according any one of claims 111-120 wherein the polymer is formed from reactants comprising a naturally occurring polymer; protein; carbohydrate; biodegradable polymer; nonbiodegradable polymer; collagen; methylated collagen; fibrinogen; thrombin; blood plasma; calcium salt; an antifibronolytic agent; fibrinogen analog; albumin; plasminogen; von Willebrands factor; factor VIII; hypoallergenic collagen; ateiopeptidic collagen; crosslinked collagen; aprotinin; epsilon-amino-n-caproic acid; gelatin; protein conjugates; gelatin conjugates; a synthetic polymer; isocyanate-containing compound; a synthetic thiol-containing compound; a synthetic compound containing at least two thiol groups; a synthetic compound containing at least three thiol groups; a synthetic compound containing at least four thiol groups; a synthetic amino-containing compound; a synthetic compound containing at least two amino groups; a synthetic compound containing at least three amino groups; a synthetic compound containing at least four amino groups; a synthetic compound comprising a carbonyl-oxygen-succinimidyl group; a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups; a synthetic polyalkylene oxide-containing compound; a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks; a synthetic polyalkylene oxide-containing compound having reactive amino groups; a synthetic polyalkylene oxide-containing compound having reactive thiol groups; a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising a biodegradable polyester block; a synthetic polymer formed in whole or part from lactic acid or lactide; a synthetic polymer formed in whole or part from glycolic acid or glycolide; polylysine; (a) protein and (b) a compound comprising a polyalkylene oxide portion; (a) protein and (b) polylysine; (a) protein and (b) a compound having at least four thiol groups; (a) protein and (b) a compound having at least four amino groups; (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) collagen and (b) a compound comprising a polyalkylene oxide portion; (a) collagen and (b) polylysine; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four amino groups; (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) cottagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion; (a) methylated collagen and (b) polylysine; (a) methylated collagen and (b) a compound having at least four thiol groups; (a) methylated collagen and (b) a compound having at least four amino groups; (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen- succinimide groups; (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; hyaluronic acid; a hyaluronic acid derivative; pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000; pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000; or (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
125. The method for implanting a medical device according any one of claims 111-120 wherein the composition comprises a colorant.
PCT/US2006/011726 2005-05-10 2006-03-31 Implantable sensors and pumps, anti-scarring agents, and therapeutic compositions WO2006121522A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US67929105P 2005-05-10 2005-05-10
US67929305P 2005-05-10 2005-05-10
US60/679,293 2005-05-10
US60/679,291 2005-05-10

Publications (2)

Publication Number Publication Date
WO2006121522A2 true WO2006121522A2 (en) 2006-11-16
WO2006121522A3 WO2006121522A3 (en) 2008-05-02

Family

ID=37397029

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/011726 WO2006121522A2 (en) 2005-05-10 2006-03-31 Implantable sensors and pumps, anti-scarring agents, and therapeutic compositions

Country Status (1)

Country Link
WO (1) WO2006121522A2 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8101371B2 (en) 2007-10-18 2012-01-24 Musc Foundation For Research Development Methods for the diagnosis of genitourinary cancer
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10583308B2 (en) 2009-06-01 2020-03-10 Profusa, Inc. Method and system for directing a localized biological response to an implant
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4871351A (en) * 1984-09-28 1989-10-03 Vladimir Feingold Implantable medication infusion system
US20040018228A1 (en) * 2000-11-06 2004-01-29 Afmedica, Inc. Compositions and methods for reducing scar tissue formation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4871351A (en) * 1984-09-28 1989-10-03 Vladimir Feingold Implantable medication infusion system
US20040018228A1 (en) * 2000-11-06 2004-01-29 Afmedica, Inc. Compositions and methods for reducing scar tissue formation

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8101371B2 (en) 2007-10-18 2012-01-24 Musc Foundation For Research Development Methods for the diagnosis of genitourinary cancer
US10583308B2 (en) 2009-06-01 2020-03-10 Profusa, Inc. Method and system for directing a localized biological response to an implant
EP2437846B1 (en) * 2009-06-01 2021-06-23 Profusa, Inc. Method and system for directing a localized biological response to an implant
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9255108B2 (en) 2012-04-10 2016-02-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10329299B2 (en) 2013-10-04 2019-06-25 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11541059B2 (en) 2014-03-19 2023-01-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10675286B2 (en) 2014-03-19 2020-06-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10941162B2 (en) 2014-10-03 2021-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10253047B2 (en) 2014-10-03 2019-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11247995B2 (en) 2015-09-14 2022-02-15 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof

Also Published As

Publication number Publication date
WO2006121522A3 (en) 2008-05-02

Similar Documents

Publication Publication Date Title
US20090280153A1 (en) electrical devices, anti-scarring agents, and therapeutic compositions
US20100092536A1 (en) Implantable sensors and implantable pumps and anti-scarring agents
WO2006121522A2 (en) Implantable sensors and pumps, anti-scarring agents, and therapeutic compositions
US20050187140A1 (en) Polymer compositions and methods for their use
US20050209664A1 (en) Electrical devices and anti-scarring agents
WO2006135479A2 (en) Anti-scarring agents, therapeutic compositions, and use thereof
CN101420970A (en) Implantable sensors and implantable pumps and anti-scarring agents
WO2006121521A2 (en) Soft tissue implants, anti-scarring agents, and therapeutic compositions

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

NENP Non-entry into the national phase in:

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06748963

Country of ref document: EP

Kind code of ref document: A2