WO2001068158A1 - Coating that promotes endothelial cell adherence - Google Patents

Coating that promotes endothelial cell adherence Download PDF

Info

Publication number
WO2001068158A1
WO2001068158A1 PCT/US2001/008244 US0108244W WO0168158A1 WO 2001068158 A1 WO2001068158 A1 WO 2001068158A1 US 0108244 W US0108244 W US 0108244W WO 0168158 A1 WO0168158 A1 WO 0168158A1
Authority
WO
WIPO (PCT)
Prior art keywords
medical device
matrix
antibody
endothelial cell
monoclonal antibody
Prior art date
Application number
PCT/US2001/008244
Other languages
French (fr)
Inventor
Michael John Bradley Kutryk
Robert John Cottone, Jr.
Stephen Maxwell Rowland
Original Assignee
Orbus Medical Technologies Inc.
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26885401&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2001068158(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Orbus Medical Technologies Inc. filed Critical Orbus Medical Technologies Inc.
Priority to CA002400319A priority Critical patent/CA2400319C/en
Priority to KR1020027012043A priority patent/KR100860860B1/en
Priority to JP2001566720A priority patent/JP5859179B2/en
Priority to IL15150101A priority patent/IL151501A0/en
Priority to EP01918685A priority patent/EP1263484B1/en
Priority to DE60128451T priority patent/DE60128451T2/en
Priority to AU2001245734A priority patent/AU2001245734A1/en
Publication of WO2001068158A1 publication Critical patent/WO2001068158A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/04Hollow or tubular parts of organs, e.g. bladders, tracheae, bronchi or bile ducts
    • A61F2/06Blood vessels
    • A61F2/07Stent-grafts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/24Heart valves ; Vascular valves, e.g. venous valves; Heart implants, e.g. passive devices for improving the function of the native valve or the heart muscle; Transmyocardial revascularisation [TMR] devices; Valves implantable in the body
    • A61F2/2412Heart valves ; Vascular valves, e.g. venous valves; Heart implants, e.g. passive devices for improving the function of the native valve or the heart muscle; Transmyocardial revascularisation [TMR] devices; Valves implantable in the body with soft flexible valve members, e.g. tissue valves shaped like natural valves
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/82Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • A61K38/363Fibrinogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/02Inorganic materials
    • A61L27/08Carbon ; Graphite
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/22Polypeptides or derivatives thereof, e.g. degradation products
    • A61L27/227Other specific proteins or polypeptides not covered by A61L27/222, A61L27/225 or A61L27/24
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/28Materials for coating prostheses
    • A61L27/30Inorganic materials
    • A61L27/303Carbon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L27/44Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L29/00Materials for catheters, medical tubing, cannulae, or endoscopes or for coating catheters
    • A61L29/08Materials for coatings
    • A61L29/10Inorganic materials
    • A61L29/103Carbon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L29/00Materials for catheters, medical tubing, cannulae, or endoscopes or for coating catheters
    • A61L29/12Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L29/126Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L29/00Materials for catheters, medical tubing, cannulae, or endoscopes or for coating catheters
    • A61L29/14Materials characterised by their function or physical properties, e.g. lubricating compositions
    • A61L29/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/02Inorganic materials
    • A61L31/022Metals or alloys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/04Macromolecular materials
    • A61L31/043Proteins; Polypeptides; Degradation products thereof
    • A61L31/047Other specific proteins or polypeptides not covered by A61L31/044 - A61L31/046
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/082Inorganic materials
    • A61L31/084Carbon; Graphite
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/12Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L31/125Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/148Materials at least partially resorbable by the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/069Vascular Endothelial cells
    • C12N5/0692Stem cells; Progenitor cells; Precursor cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/20Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices containing or releasing organic materials
    • A61L2300/252Polypeptides, proteins, e.g. glycoproteins, lipoproteins, cytokines
    • A61L2300/256Antibodies, e.g. immunoglobulins, vaccines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/422Anti-atherosclerotic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/432Inhibitors, antagonists
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/606Coatings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2420/00Materials or methods for coatings medical devices
    • A61L2420/04Coatings containing a composite material such as inorganic/organic, i.e. material comprising different phases
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to the field of medical devices implanted in vessels within the body More particularly, the present invention relates to stents or synthetic grafts implanted in blood vessels that incorporate a mat ⁇ x which promotes adherence of endothelial cells to the stent or synthetic graft
  • Atherosclerosis is one of the leading causes of death and disability in the world Atherosclerosis involves the deposition of fatty plaques on the lumenal surface of arteries The deposition of fatty plaques on the lumenal surface of the artery causes narrowing of the cross- sectional area of the artery Ultimately, this deposition blocks blood flow distal to the lesion causing ischemic damage to the tissues supplied by the artery
  • Coronary artifacts supply the heart with blood Coronary artery atherosclerosis disease is the most common, serious, chronic, life-threatening illness in the United States, affecting more than 11 million persons
  • the social and economic costs of coronary atherosclerosis vastly exceed that of most other diseases
  • Narrowing of the coronary artery lumen causes destruction of heart muscle resulting first in angina, followed by myocardial infarction and finally death
  • myocardial infarctions There are over 1 5 million myocardial infarctions in the United States each year Six hundred thousand (or 40%) of those patients suffer an acute myocardial infarction and more than three hundred thousand of those patients die before reaching the hospital (Harrison's Principles of Internal Medicine.14 th Edition. 1998)
  • CAD can be treated using percutaneous translumenal coronary balloon angioplasty (PTCA) More than 400,000 PTCA procedures are performed each year in the United States
  • PTCA percutaneous translumenal coronary balloon angioplasty
  • a balloon catheter is inserted into a peripheral artery and threaded through the arterial system into the blocked coronary artery
  • the balloon is then inflated, the artery stretched, and the obstructing fatty plaque flattened, thereby increasing the cross-sectional flow of blood through the affected artery
  • the therapy does not usually result in a permanent opening of the affected coronary artery As many as 50% of the patients who are treated by PTCA require a repeat procedure within six months to correct a re-narrowing of the coronary artery.
  • restenosis this re-narrowing of the artery after treatment by PTCA is called restenosis.
  • Acutely, restenosis involves recoil and shrinkage of the vessel.
  • recoil and shrinkage of the vessel are followed by proliferation of medial smooth muscle cells in response to injury of the artery from PTCA
  • proliferation of smooth muscle cells is mediated by release of various inflammatory factors from the injured area including thromboxane A 2> platelet derived growth factor (PDGF) and fibroblast growth factor (FGF).
  • PDGF platelet derived growth factor
  • FGF fibroblast growth factor
  • Stents have proven to be the most effective Stents are metal scaffolds that are positioned in the diseased vessel segment to create a normal vessel lumen Placement of the stent in the affected arterial segment prevents recoil and subsequent closing of the artery. Stents can also prevent local dissection of the artery along the medial layer of the artery By maintaining a larger lumen than that created using PTCA alone, stents reduce restenosis by as much as 30%. Despite their success, stents have not eliminated restenosis entirely (Suryapranata et al 1998 Randomized comparison of coronary stenting with balloon angioplasty in selected patients with acute myocardial infarction. Circulation 97:2502-2502).
  • Narrowing of the arteries can occur in vessels other than the coronary arteries, including the aortoiliac, infrainguinal, distal profunda femoris, distal popliteal, tibial, subclavian and mesenteric arteries.
  • the prevalence of peripheral artery atherosclerosis disease (PAD) depends on the particular anatomic site affected as well as the criteria used for diagnosis of the occlusion. Traditionally, physicians have used the test of intermittent claudication to determine whether PAD is present.
  • PAD can be treated using percutaneous translumenal balloon angioplasty (PTA).
  • PTA percutaneous translumenal balloon angioplasty
  • the use of stents in conjunction with PTA decreases the incidence of restenosis.
  • the post-operative results obtained with medical devices such as stents do not match the results obtained using standard operative revascularization procedures, i.e., those using a venous or prosthetic bypass material.
  • PAD is treated using bypass procedures where the blocked section of the artery is bypassed using a graft.
  • the graft can consist of an autologous venous segment such as the saphenous vein or a synthetic graft such as one made of polyester, polytetrafluoroethylene (PTFE), or expanded polytetrafluoroethylene (ePTFE).
  • PTFE polytetrafluoroethylene
  • ePTFE expanded polytetrafluoroethylene
  • the post-operative patency rates depend on a number of different factors, including the lumenal dimensions of the bypass graft, the type of synthetic material used for the graft and the site of outflow. Restenosis and thrombosis, however, remain significant problems even with the use of bypass grafts. For example, the patency of infrainguinal bypass procedures at 3 years using an ePTFE bypass graft is 54% for a femoral-popliteal bypass and only 12% for a femoral-tibial bypass.
  • stents With stents, the approach has been to coat the stents with various anti-thrombotic or anti-restenotic agents in order to reduce thrombosis and restenosis.
  • impregnating stents with radioactive material appears to inhibit restenosis by inhibiting migration and proliferation of myofibroblasts.
  • Irradiation of the treated vessel can pose safety problems for the physician and the patient.
  • irradiation does not permit uniform treatment of the affected vessel.
  • stents have also been coated with chemical agents such as heparin or phosphorylcholine, both of which appear to decrease thrombosis and restenosis.
  • heparin and phosphorylcholine appear to markedly reduce restenosis in animal models in the short term, treatment with these agents appears to have no long-term effect on preventing restenosis. Additionally, heparin can induce thrombocytopenia, leading to severe thromboembolic complications such as stroke. Nonetheless, it is not feasible to load stents with sufficient therapeutically effective quantities of either heparin or phosphorylcholine to make treatment of restenosis in this manner practical.
  • Synthetic grafts have been treated in a variety of ways to reduce postoperative restenosis and thrombosis. (Bos et al. 1998. Small-Diameter Vascular Graft Prostheses:Current Status Archives Physio. Biochem. 106: 100-115). For example, composites of polyurethane such as meshed polycarbonate urethane have been reported to reduce restenosis as compared with ePTFE grafts. The surface of the graft has also been modified using radiofrequency glow discharge to add polyterephalate to the ePTFE graft. Synthetic grafts have also been impregnated with biomolecules such as collagen. However, none of these approaches has significantly reduced the incidence of thrombosis or restenosis over an extended period of time.
  • endothelial cells possess certain intrinsic characteristics such as cell regulatory molecules that decrease the incidence of thrombosis or restenosis, stimulating the development of an endothelial cell monolayer on the surface of stents or synthetic grafts may prevent both restenosis and thrombosis.
  • endothelial cells possess certain intrinsic characteristics such as cell regulatory molecules that decrease the incidence of thrombosis or restenosis
  • stimulating the development of an endothelial cell monolayer on the surface of stents or synthetic grafts may prevent both restenosis and thrombosis.
  • VEGF vascular endothelial growth factor
  • Synthetic grafts have also been seeded with endothelial cells, but the clinical results with endothelial seeding have been generally poor, i.e., low post-operative patency rates (Lio et al. 1998. New concepts and Materials in Microvascular Grafting: Prosthetic Graft Endothelial Cell Seeding and Gene Therapy. Microsurgery 18:263-256).
  • the invention provides methods and compositions for coating medical devices with a matrix that promotes adherence of endothelial cells to a medical device.
  • the matrix incorporates antibodies that stimulate adherence of endothelial cells to the surface of the medical device.
  • medical device refers to a device that is introduced temporarily or permanently into a mammal for the prophylaxis or therapy of a medical condition. These devices include any that are introduced subcutaneously, percutaneously or surgically to rest within an organ, tissue or lumen.
  • Medical devices may include stents, covered stents such as those covered with polytetrafluoroethylene (PTFE), or expanded polytetrafluoroethylene (ePTFE), synthetic grafts, artificial heart valves, artificial hearts and fixtures to connect the prosthetic organ to the vascular circulation, venous valves, abdominal aortic aneurysm (AAA) grafts, inferior venal caval filters, permanent drug infusion catheters, embolic coils, embolic materials used in vascular embolization (e.g., PVA foams), and vascular sutures.
  • PTFE polytetrafluoroethylene
  • ePTFE expanded polytetrafluoroethylene
  • Coating of the medical device with the compositions and methods of this invention may stimulate the development of an endothelial cell layer on the surface of the medical device, thereby preventing restenosis as well as other thromboembolic complications that result from implantation of the medical device.
  • Synthetic grafts and stents can be used for treating CAD or PAD.
  • a stent or synthetic graft may be coated with a matrix incorporating antibodies that stimulate adherence of circulating progenitor endothelial cells to the medical device.
  • the antibodies may comprise monoclonal antibodies reactive with endothelial cell surface antigens such as CD34, an antigen expressed on the surface of progenitor endothelial cells.
  • Fab fragments of the monoclonal antibody may be used.
  • monoclonal antibodies directed against other endothelial surface antigens such as KDR or Tie-2, may also be used.
  • a single type of antibody that reacts with one antigen may be used.
  • a plurality of different antibodies directed against different endothelial cell surface antigens may be mixed together and added to the matrix.
  • the matrix coating the medical device may be composed of synthetic material, such as polyurethane, poly-L-lactic acid, cellulose ester or polyethylene glycol.
  • the matrix is composed of naturally occurring materials, such as collagen, fibrin, elastin or amorphous carbon.
  • the matrix may comprise several layers with a first layer being composed of synthetic or naturally occurring materials and a second layer composed of antibodies. The layers may be ordered sequentially, with the first layer directly in contact with the stent or synthetic graft surface and the second layer having one surface in contact with the first layer and the opposite surface in contact with the vessel lumen.
  • the matrix may comprise fullerenes, where the fullerenes range from about C60 to about CIOO.
  • the fullerenes may also be arranged as nanotubes, that incorporate molecules or proteins.
  • the fullerene matrix may also be mixed with PTFE or ePTFE, or antibodies.
  • the PTFE or ePTFE may be layered first on the medical device followed by a second layer of fullerenes.
  • the matrix may be noncovalently or covalently attached to the medical device.
  • Antibodies may be covalently attached to the matrix using hetero- or homobifunctional cross- linking reagents.
  • the artery may be the either a coronary artery or a peripheral artery such as the femoral artery.
  • Figure 1 shows an antibody tethered covalently to the matrix by a cross-linking molecule
  • Figure 2 shows a diagram of the C60O molecule anchoring the matnx DETAILED DESCRIPTION OF THE INVENTION
  • the present invention provides methods and compositions that involve coating a medical device such as a stent or synthetic graft with a matrix which is then used to coat the medical device
  • the matrix incorporates a therapeutically effective amount of at least one type of antibody that promotes adherence of endothelial cells to the medical device Following adherence, the endothelial cells differentiate and proliferate on the surface of the matrix
  • the presence of endothelial cells on the medical device reduces the occurrence of restenosis and thrombosis after medical device implantation into a vessel
  • antibody refers to one type of monoclonal or polyclonal antibody, where the monoclonal or polyclonal antibody binds to one antigen or a functional equivalent of that antigen
  • antibody encompasses any fragment of an antibody such as Fab, F(ab') 2 or Fc fragments (An antibody encompasses a plurality of individual antibody molecules equal to 6 022 x 10 23 molecules per mole of antibody)
  • a "therapeutically effective amount of the antibody” means the amount of an antibody that promotes adherence of endothelial cells to the medical device.
  • the amount of an antibody needed to practice the claimed invention varies with the nature of the antibody used For example, the amount of an antibody used will depend on the binding constant between the antibody and the antigen against which it reacts It is well known to those of ordinary skill in the art how to determine therapeutically effective amounts of an antibody to use with a particular antigen.
  • medical device refers to a device that is introduced temporarily or permanently into a mammal for the prophylaxis or therapy of a medical condition
  • These devices include any that are introduced subcutaneously, percutaneously or surgically to rest within an organ, tissue or lumen
  • Medical devices may include, stents, covered stents such as those covered with PTFE, or ePTFE, synthetic grafts, artificial heart valves, artificial hearts and fixtures to connect the prosthetic organ to the vascular circulation, venous valves, abdominal aortic aneurysm (AAA) grafts, infenor venal caval filters, permanent drug infusion catheters, embolic coils, embolic materials used in vascular embolization (e g , PVA foams), and vascular sutures
  • restenosis refers to the accumulation of a layer of smooth muscle cells and matrix protein in the intima of an arterial wall Vessels may become obstructed because of restenosis.
  • smooth muscle cells from the media and adventitia which are not normally present in the intima, proliferate and migrate to the intima and secrete proteins, forming an accumulation of smooth muscle cells and matrix protein within the intima. This accumulation causes a narrowing of the lumen of the artery, reducing blood flow distal to the narrowing.
  • inhibittion of restenosis refers to the inhibition of migration and proliferation of smooth muscle cells accompanied by prevention of protein secretion so as to prevent restenosis and the complications arising therefrom.
  • the subjects that can be treated using the methods and compositions of this invention may be a mammal, or more specifically, a human, dog, cat, pig, rodent or monkey.
  • the methods of the present invention may be practiced in vivo or in vitro.
  • endothelial cell refers to endothelial cells at any developmental stage, from progenitor to mature. Fully differentiated endothelial cells may be isolated from an artery or vein such as a human umbilical vein, while progenitor endothelial cells are isolated from peripheral blood or bone marrow. The endothelial cells are bound to the medical devices by incubation of the endothelial cells with a medical device coated with the matrix that incorporates an antibody or other agent that adheres to endothelial cells.
  • the methods of this invention may be practiced on any artery or vein. Included within the scope of this invention is atherosclerosis of any artery including coronary, infrainguinal, aortoiliac, subclavian, mesenteric and renal arteries. Other types of vessel obstructions, such as those resulting from a dissecting aneurysm are also encompassed by the invention.
  • the medical device may be coated with endothelial cells after insertion into a vessel.
  • the medical device is coated with the endothelial cells before insertion of the medical device.
  • the presence of endothelial cells on the lumenal surface of the medical device inhibits or prevents restenosis and thrombosis.
  • Human umbilical vein endothelial cells are obtained from umbilical cords according to the methods of Jaffe, et al., J. Clin. Invest.. 52:2745-2757, 1973, incorporated herein by reference. Briefly, cells are stripped from the blood vessel walls by treatment with collagenase and cultured in gelatin-coated tissue culture flasks in Ml 99 medium containing 10% low endotoxin fetal calf serum, 90 ug/ml preservative-free porcine heparin, 20 ug/ml endothelial cell growth supplement (ECGS), glutamine and antibodies.
  • ECGS endothelial cell growth supplement
  • Progenitor endothelial cells are isolated from human peripheral blood according to the methods of Asahara et al. (Isolation of putative progenitor endothelial cells for angiogenesis. Science 275:964-967. 1997, incorporated herein by reference). Magnetic beads coated with antibody to CD34 are incubated with human peripheral blood. After incubation, bound cells are eluted and can be cultured in M- 199 containing 20% fetal bovine serum and bovine brain extract. (Clonetics, San Diego, CA). Cells are characterized by fluorescent antibodies to CD45, CD34, CD31, Flk-1, Tie-2, and E-selectin.
  • Endothelial cells are transfected with any mammalian expression vectors that contains any cloned genes encoding proteins such as platelet derived growth factor (PDGF), fibroblast growth factor (FGF), or nitric oxide synthase (NOS) using conventional methods.
  • PDGF platelet derived growth factor
  • FGF fibroblast growth factor
  • NOS nitric oxide synthase
  • mammalian expression vectors and transfection kits commercially available from Stratagene, San Diego, CA.
  • purified porcine progenitor endothelial cells are transfected with vascular endothelial growth factor (VEGF) using an adenoviral expression vector expressing the VEGF cDNA according to the methods of Rosengart et al.
  • VEGF vascular endothelial growth factor
  • Monoclonal antibodies useful in the method of the invention may be produced according to the standard techniques of Kohler and Milstein (Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 265:495-497, 1975, incorporated herein by reference). Endothelial cells can be used as the immunogen to produce monoclonal antibodies directed against endothelial cell surface antigens.
  • Monoclonal antibodies directed against endothelial cells are prepared by injecting HUVEC or purified progenitor endothelial cells into a mouse or rat. After a sufficient time, the mouse is sacrificed and spleen cells are obtained. The spleen cells are immortalized by fusing them with myeloma cells or with lymphoma cells, generally in the presence of a non-ionic detergent, for example, polyethylene glycol. The resulting cells, which include the fused hybridomas, are allowed to grow in a selective medium, such as HAT-medium, and the surviving cells are grown in such medium using limiting dilution conditions. The cells are grown in a suitable container, e.g., microtiter wells, and the supernatant is screened for monoclonal antibodies having the desired specificity, i.e , reactivity with endothelial cell antigens
  • useful binding fragments of anti-endothelial cell monoclonal antibodies such as the Fab, F(ab') 2 , or Fc fragments of these monoclonal antibodies.
  • the antibody fragments are obtained by conventional techniques.
  • useful binding fragments may be prepared by peptidase digestion of the antibody using papain or pepsin.
  • Antibodies of the invention are directed to an antibody of the IgG class from a murine source; however, this is not meant to be a limitation.
  • the above antibody and those antibodies having functional equivalency with the above antibody, whether from a murine source, mammalian source including human, or other sources, or combinations thereof are included within the scope of this invention, as well as other classes such as IgM, IgA, IgE, and the like, including isotypes within such classes.
  • the term "functional equivalency" means that two different antibodies each bind to the same antigenic site on an antigen, in other words, the antibodies compete for binding to the same antigen.
  • the antigen may be on the same or different molecule.
  • monoclonal antibodies reacting with the endothelial cell surface antigen CD34 are used.
  • Anti-CD34 monoclonal antibodies attached to a solid support have been shown to capture progenitor endothelial cells from human peripheral blood. After capture, these progenitor cells are capable of differentiating into endothelial cells.
  • Hybridomas producing monoclonal antibodies directed against CD34 can be obtained from the American Type Tissue Collection. (Rockville, MD).
  • monoclonal antibodies reactive with endothelial cell surface antigens Flk-1 or Tie-2 are used. Polyclonal antibodies reactive against endothelial cells isolated from the same species as the one receiving the medical device implant may also be used.
  • Stent herein means any medical device which when inserted into the lumen of a vessel expands the cross-sectional lumen of a vessel.
  • the term “stent” includes covered stents such as those covered with PTFE or ePTFE. In one embodiment, this includes stents delivered percutaneously to treat coronary artery occlusions or to seal dissections or aneurysms of the splenic, carotid, iliac and popliteal vessels. In another embodiment, the stent is delivered into a venous vessel.
  • the stent can be composed of polymeric or metallic structural elements onto which the matrix is applied or the stent can be a composite of the matrix intermixed with a polymer.
  • a deformable metal wire stent can be used, such as that disclosed in U.S. Pat. No. 4,886,062 to Wiktor, incorporated herein by reference.
  • a self-expanding stent of resilient polymeric material such as that disclosed in published international patent application WO91/12779 "Intraluminal Drug Eluting Prosthesis", incorporated herein by reference, can also be used.
  • Stents may also be manufactured using stainless steel, polymers, nickel-titanium, tantalum, gold, platinum-iridium, or Elgiloy and MP35N and other ferrous materials.
  • Stents are delivered through the body lumen on a catheter to the treatment site where the stent is released from the catheter, allowing the stent to expand into direct contact with the lumenal wall of the vessel. It will be apparent to those skilled in the art that other self-expanding stent designs (such as resilient metal stent designs) could be used with the antibodies and matrices of this invention.
  • Synthetic Graft means any artificial prosthesis having biocompatible characteristics. In one embodiment this includes synthetic grafts made of Dacron (polyethylene terephthalate, PET) or Teflon (ePTFE). In another embodiment, synthetic grafts are composed of polyurethane. In yet a third embodiment, a synthetic graft is composed of an inner layer of meshed polycarbonate urethane and an outer layer of meshed Dacron. It will be apparent to those skilled in the art that any biocompatible synthetic graft can be used with the antibodies and matrices of this invention. (Bos et al. 1998. Small-Diameter Vascular Prostheses: Current Status. Archives Physio Biochem. 106:100-115, incorporated herein by reference). Synthetic grafts can be used for end-to-end anastomosis of vessels or for bypass of a diseased vessel segment.
  • the matrix that is used to coat the stent or synthetic graft may be selected from synthetic materials such as polyurethane, segmented polyurethane-urea/heparin, poly-L-lactic acid, cellulose ester or polyethylene glycol.
  • Naturally Occurring Material The matrix may be selected from naturally occurring substances such as collagen, laminin, heparin, fibrin, cellulose or carbon. A primary requirement for the matrix is that it be sufficiently elastic and flexible to remain unruptured on the exposed surfaces of the stent or synthetic graft.
  • C Fullerenes - The matrix may also comprise a fullerene (the term "fullerene” encompasses a plurality of fullerene molecules). Fullerenes are carbon-cage molecules. The number of carbon
  • (C) molecules in a fullerene species varies from about C60 to about CIOO.
  • Fullerenes are produced by high temperature reactions of elemental carbon or of carbon-containing species by processes well known to those skilled in the art; for example, by laser vaporization of carbon, heating carbon in an electric arc or burning of hydrocarbons in sooting flames.
  • U.S. Patent No. 5,292,813, to Patel et al., incorporated herein by reference; U.S. Patent No. 5,558,903 to Bhushan et al., incorporated herein by reference In each case, a carbonaceous deposit or soot is produced. From this soot, various fullerenes are obtained by extraction with appropriate solvents, such as toluene.
  • fullerenes are separated by known methods, in particular by high performance liquid chromatography (HPLC). Fullerenes may be synthesized or obtained commercially from Dynamic Enterprises, Ltd., Berkshire, England or Southern Chemical Group, LLC, Tucker, Georgia.
  • HPLC high performance liquid chromatography
  • Fullerenes may be deposited on surfaces in a variety of different ways, including, sublimation, laser vaporization, sputtering, ion beam, spray coating, dip coating, roll-on or brush coating as disclosed in U.S. Patent No. 5,558,903.
  • fullerenes An important feature of fullerenes is their ability to form "activated carbon.”
  • the fullerene electronic structure is a system of overlapping pi-orbitals, such that a multitude of bonding electrons are cooperatively presented around the surface of the molecule. (Chemical and Engineering News. Apr. 8, 1991, page 59, incorporated herein by reference).
  • As forms of activated carbon fullerenes exhibit substantial van der Waals forces for weak interactions.
  • the adsorptive nature of the fullerene surface may lend itself to additional modifications for the purpose of directing specific cell membrane interactions.
  • lectins or antibodies can be adsorbed to the fullerene surface.
  • the fullerene surface may also be chemically modified to present specifically reactive groups to the cell membrane, e.g., oxidants or reductants. Attachment of different molecules to the fullerene surface may be manipulated to create surfaces that selectively bind various cell types, e.g., epithelial cells, fibroblasts, primary explants, or T-cell subpopulations.
  • Fullerenes may also form nanotubes that incorporate other atoms or molecules.
  • the synthesis and preparation of carbon nanotubes is well known in the art.
  • Molecules such as proteins may also be incorporated inside carbon nanotubes.
  • nanotubes may be filled with the enzymes, e.g., Zn 2 Cd -metallothionein, cytochromes C and C3, and beta-lactamase after cutting the ends of the nanotube.
  • enzymes e.g., Zn 2 Cd -metallothionein, cytochromes C and C3, and beta-lactamase after cutting the ends of the nanotube.
  • U.S. Patent No. 5,338,571 to Mirkin et al. discloses three-dimensional, multilayer fullerene structures that are formed on a substrate surface by (i) chemically modifying fullerenes to provide a bond-forming species; (ii) chemically treating a surface of the substrate to provide a bond-forming species effective to covalently bond with the bond-forming species of the fullerenes in solution; and, (iii) contacting a solution of modified fullerenes with the treated substrate surface to form a fullerene layer covalently bonded to the treated substrate surface.
  • the matrix should adhere tightly to the surface of the stent or synthetic graft. Preferably, this is accomplished by applying the matrix in successive thin layers.
  • Each layer of matrix may incorporate the antibodies.
  • antibodies may be applied only to the layer in direct contact with the vessel lumen.
  • Different types of matrices may be applied successively in succeeding layers.
  • the antibodies may be covalently or noncovalently coated on the matrix after application of the matrix to the stent.
  • the stent In order to coat a medical device such as a stent, the stent is dipped or sprayed with a liquid solution of the matrix of moderate viscosity. After each layer is applied, the stent is dried before application of the next layer.
  • a thin, paint-like matrix coating does not exceed an overall thickness of 100 microns.
  • a suitable matrix coating solution is prepared by dissolving 480 milligrams (mg) of a drug carrier, such as poly-D, L-lactid (available as R203 of Boehringer Inc., Ingelheim, Germany) in 3 milliliters (ml) of chloroform under aseptic conditions.
  • a drug carrier such as poly-D, L-lactid (available as R203 of Boehringer Inc., Ingelheim, Germany) in 3 milliliters (ml) of chloroform under aseptic conditions.
  • any biodegradable (or non-biodegradable) matrix that is blood-and tissue-compatible (biocompatible) and can be dissolved, dispersed or emulsified may be used as the matrix if, after application, it undergoes relatively rapid drying to a self-adhesive lacquer- or paint-like coating on the medical device.
  • fibrin is clotted by contacting fibrinogen with thrombin.
  • the fibrin in the fibrin- containing stent of the present invention has Factor XIII and calcium present during clotting, as described in U.S. Pat. No. 3,523,807 issued to Gerendas, incorporated herein by reference, or as described in published European Patent Application 0366564, incorporated herein by reference, in order to improve the mechanical properties and biostability of the implanted device.
  • the fibrinogen and thrombin used to make fibrin in the present invention are from the same animal or human species as that in which the stent will be implanted in order to avoid any inter-species immune reactions, e g., human anti-cow.
  • the fibrin product can be in the form of a fine, fibrin film produced by casting the combined fibrinogen and thrombin in a film and then removing moisture from the film osmotically through a semipermeable membrane.
  • a substrate preferably having high porosity or high affinity for either thrombin or fibrinogen
  • a fibrinogen solution is contacted with a fibrinogen solution and with a thrombin solution.
  • fibrin layer formed by polymerization of fibrinogen on the surface of the medical device.
  • Multiple layers of fibrin applied by this method could provide a fibrin layer of any desired thickness.
  • the fibrin can first be clotted and then ground into a powder which is mixed with water and stamped into a desired shape in a heated mold (U.S. Patent No. 3,523,807).
  • Increased stability can also be achieved in the shaped fibrin by contacting the fibrin with a fixing agent such as glutaraldehyde or formaldehyde.
  • a synthetic graft is coated with collagen
  • the methods for preparing collagen and forming it on synthetic graft devices are well known as set forth in U.S. Patent No. 5,851,230 to Weadock et al., incorporated herein by reference.
  • This patent describes methods for coating a synthetic graft with collagen.
  • Methods for adhering collagen to a porous graft substrate typically include applying a collagen dispersion to the substrate, allowing it to dry and repeating the process.
  • Collagen dispersions are typically made by blending insoluble collagen (approximately 1- 2% by weight) in a dispersion at acidic pH (a pH in a range of 2 to 4).
  • the dispersion is typically injected via syringe into the lumen of a graft and massaged manually to cover the entire inner surface area with the collagen slurry. Excess collagen slurry is removed through one of the open ends of the graft Coating and drying steps are repeated several times to provide sufficient treatment
  • the stent or synthetic graft is coated with amorphous carbon
  • a method for producing a high-rate, low-temperature deposition of amorphous carbon films in the presence of a fluo ⁇ nated or other halide gas is descnbed
  • Deposition according to the methods of this invention can be performed at less than 100° C, including ambient room temperature, with a radio- frequency, plasma-assisted, chemical-vapor deposition process
  • the amorphous carbon film produced using the methods of this invention adheres well to many types of substrates, including for example glasses, metals, semiconductors, and plastics
  • amine-containing polymers may be performed as described in U S Patent No 5,292,813 Chemical modification in this manner allows for direct incorporation of the fullerenes into the stent
  • the fullerenes may be deposited on the surface of the stent or synthetic grafts as described above (see, WO 99/32184 to Leone et al , incorporated by reference)
  • Fullerenes may also be attached through an aldehyde bond (Yamago et al , Chemical Denvatization of Organofullerenes through Oxidation, Reduction and C-0 and C-C Bond Forming Reactions J Org Chem .
  • C60O may also be attached directly through an epoxide group on the fullerene to a stent The attachment is through a covalent linkage to the oxygen
  • This compound and the protocols for coupling are commercially available from BuckyUSA (BuckyUSA, Houston, Texas)
  • the antibodies are added to a solution containing the mat ⁇ x
  • Fab fragments on anti-CD34 monoclonal antibody are incubated with a solution containing human fibrinogen at a concentration of between 500 and 800 mg/dl
  • concentration of anti-CD34 Fab fragment will vary and that one of ordinary skill in the art could determine the optimal concentration without undue experimentation
  • the stent is added to the Fab/fibrin mixture and the fib ⁇ n activated by addition of concentrated thrombin (at a concentration of at least lOOOU/ml)
  • the resulting polymerized fibrin mixture containing the Fab fragments incorporated directly into the matrix is pressed into a thin film (less than 0.12 cm) on the surface of the stent or synthetic graft.
  • antibodies are covalently coupled to the matrix.
  • the antibodies are tethered covalently to the matrix through the use of hetero- or homobifunctional linker molecules.
  • the term "tethered” refers to a covalent coupling of the antibody to the matrix by a linker molecule.
  • linker molecules in connection with the present invention typically involves covalently coupling the linker molecules to the matrix after it is adhered to the stent.
  • the linker molecules After covalent coupling to the matrix, the linker molecules provide the matrix with a number of functionally active groups that can be used to covalently couple one or more types of antibody.
  • Figure 1 provides an illustration of coupling via a cross-linking molecule.
  • An endothelial cell, 1.01 binds to an antibody, 1.03, by a cell surface antigen, 1.02.
  • the antibody is tethered to the matrix, 1.05-1.06, by a cross-linking molecule, 1.04.
  • the matrix, 1.05-1.06 adheres to the stent, 1.07.
  • the linker molecules may be coupled to the matrix directly (i.e., through the carboxyl groups), or through well-known coupling chemistries, such as, esterification, amidation, and acylation.
  • the linker molecule may be a di- or tri-amine functional compound that is coupled to the matrix through the direct formation of amide bonds, and provides amine-functional groups that are available for reaction with the antibodies.
  • the linker molecule could be a polyamine functional polymer such as polyethyleneimine (PEI), polyallylamine (PALLA) or polyethyleneglycol (PEG).
  • PEI polyethyleneimine
  • PALLA polyallylamine
  • PEG polyethyleneglycol
  • a variety of PEG derivatives, e.g., mPEG-succinimidyl propionate or mPEG-N-hydroxysuccinimide, together with protocols for covalent coupling, are commercially available from Shearwater Corporation, Birmingham, Alabama.
  • Antibodies may be attached to C60O fullerene layers that have been deposited directly on the surface of the stent.
  • Cross linking agents may be covalently attached to the fullerenes.
  • the antibodies are then attached to the cross-linking agent, which in turn is attached to the stent.
  • Figure 2 provides an illustration of coupling by C60O.
  • the endothelial cell, 2.01 is bound via a cell surface antigen, 2 02, to an antibody, 2 03, which in turn is bound, covalently or non-covalently to the matrix, 2 04
  • the matrix, 2 04 is coupled covalently via C60O, 2 05, to the stent, 2 06
  • R stents produced by Orbus International B.V. will be incubated with human fibrinogen (Sigma, St. Louis, MO) 500-800 mg/ml together with Fab fragments of anti-CD34 monoclonal antibody and the fibrinogen will be polymerized by the addition of 1000 units/ml of thrombin.
  • human fibrinogen Sigma, St. Louis, MO
  • the fibrin will be compressed into a thin film (less than 0.012 cm) against the R Stent.
  • the R-Stent having the thin, fibrin film containing the Fab fragments will be washed three times with phosphate-buffered saline (PBS) containing 0.5% bovine serum albumin (BSA) at room temperature.
  • PBS phosphate-buffered saline
  • BSA bovine serum albumin
  • R stents will be coated with mPEG-succinimidyl propionate (Shearwater Corporation, Birmingham, Alabama). The succinimidyl group will be reacted with the anti-CD34 monoclonal Fab fragments (Fab-PEG coated R stents) according to the manufacturer's instructions to form a stable amide linkage between the PEG derivative and the Fab fragment.
  • the fibrin-anti-CD34 Fab coated R-stents or Fab-PEG coated R stents will be incubated with isolated HUVEC or isolated progenitor endothelial cells at cellular concentrations of between 100,000 and 1 ,000,000 cells/ml in M199 containing 0.5% BSA at 37°C in a 5% CO 2 humidified atmosphere. Prior to incubation with the stent, the HUVEC or progenitor endothelial cells will be labeled with [ 3 H]-thymidine for 24 hours.
  • the stents After incubation of the labeled endothelial cells with the stents coated with fibrin and Fab anti-CD34 for between 4 and 72 hours, the stents will be removed from the solution and washed 5 times with Ml 99 containing 0.5% BSA. Bound endothelial cells will be removed by trypsinization and binding of labeled endothelial cells to the stents will be assessed by scintillation counting of [ 3 H]-thymidine. As negative control, stents coated with fibrin alone or uncoated stents will be incubated with [ 3 H]-thymidine-labeled endothelial cells. Results will be evaluated statistically using a t-test to determine differential binding. Stents coated with fibrin which incorporate monoclonal anti-CD34 Fab fragments will show significantly increased binding of endothelial cells as compared with uncoated stents.
  • R stents coated with fibrin that incorporates anti-CD34 Fab fragments will be incubated with HUVEC or progenitor endothelial cells for between 4 and 72 hours in Ml 99 containing 0.5% BSA. After incubation of the stents with the HUVEC or progenitor endothelial cells, the stents will be washed 5 times with Ml 99 containing 0.5% BSA and then incubated with [ 3 H]-thymidine. [ 3 H]-thymidine incorporation will be assessed in washed and harvested HUVEC or progenitor endothelial cells (cells will be harvested with trypsin).
  • Proliferation of HUVEC or progenitor endothelial cells on fibrin-coated stents will be compared with endothelial cell proliferation in standard microtiter dishes. Proliferation of HUVEC or progenitor endothelial cells on fibrin-coated stents will be equal to or greater than proliferation of endothelial cells in microtiter dishes
  • BALB/c mice will be immunized, intraperitoneally 3-4 times at 2-4 weekly intervals, with 1.5 x 10 6 HUVEC in PBS or 1.5 x 10 6 progenitor endothelial cells and challenged 3 days prior to spleen-cell removal with 1.5 x 10 6 HUVEC or 1.5 x 10 6 progenitor endothelial cells.
  • a spleen-cell suspension will be prepared, fused with the myeloma NS 1/1 AG4.1 and hybridomas grown up and cloned.
  • 10% endothelial-cell conditioned medium (HUVEC) will be included in culture media.
  • hybridoma culture supernatants will be tested for reactivity with HUVEC or progenitor endothelial cells by immunofluorescence flow cytometry (FACS). Briefly, HUVEC (1.5 x 10 4 ) or progenitor endothelial cells (1.5 x 10 4 ) will be incubated (30 min, 4°C) with undiluted hybridoma supernatant, washed and incubated with fluorescein-isothiocyanate (FITC)-sheep F(ab') 2 anti- mouse Ig(100 ug/ml). Following final washing, the endothelial cells will be examined for monoclonal antibody binding by FACS analysis.
  • FITC fluorescein-isothiocyanate
  • Implantation of antibody-covered stents will be performed in juvenile Yorkshire pigs weighing between 25 and 30 kg. Animal care will comply with the "Guide for the Care and Use of Laboratory Animals" (NIH publication No. 80-23, revised 1985). After an overnight fast, animals will be sedated with ketamine hydrochloride (20mg/kg). Following the induction of anesthesia with thiopental (12 mg kg) the animals will be intubated and connected to a ventilator that will administer a mixture of oxygen and nitrous oxide (1 :2 [vol/vol]). Anesthesia will be maintained with 0.5-2.5 vol% isoflurane. Antibiotic prophylaxis will be provided by an intramuscular injection of 1,000 mg of a mixture of procaine penicillin-G and benzathine penicillin-G (streptomycin).
  • an arteriotomy of the left carotid artery will be performed and a 9F- introducer sheath will be placed in the left carotid artery. All animals will be given 7,500 IU of heparin sodium and 100 mg of acetylsalicylic acid intravenously. Additional 2,500 IU boluses of heparin will be administered periodically throughout the procedure in order to maintain an activated clotting time above 300 seconds.
  • An 8F guiding catheter will be introduced through the carotid sheath and passed to the origin of the iliac artery. Angiography will be performed after the administration of lmg of isosorbide dinitrate and images will be analyzed using a quantitative coronary angiography system.
  • a 3F-embolectomy catheter will be inserted into the common femoral artery, and passed distal to the segment selected for stent implantation.
  • the embolectomy balloon will be inflated to a size 0.5 mm larger than the arterial segment and withdrawn twice to denude the vessel.
  • a fibrin-coated stent incorporating an Fab fragment of a monoclonal antibody will be inserted through the guiding catheter and deployed in the denuded segment of the femoral artery.
  • Animals will be sacrificed both at 3 days and at eight weeks after stent implantation. The animal will first be sedated and anesthetized as described above.
  • the stented femoral segments will be explanted and then placed in 4% paraformaldehyde in 0.1 M phosphate buffer pH 7.2 at 4° C for 48 h.
  • a rectangular section of the vessel wall will be removed for further processing for electron microscopic evaluation of surface coverage of endothelial cells.
  • This portion of the stented vessel will be placed in 0.15 cacodylate buffer and further fixed with 2.5% glutaraldehyde in 0.15 M cacodylate.
  • the tissue will then be post-fixed with 0.1 M cacodylate buffer containing 1% OsO and 50 mM ferricya ⁇ ide (K 3 [Fe(CN)6]), and further processed.
  • Embedded arterial segments with the stent in place will be cut into sections 3 to 5 ⁇ m thick on a motor-driven rotary microtome (HM-350, Microm GmbH, Kunststoff, Germany) using stainless steel disposable knives
  • HM-350 motor-driven rotary microtome
  • sections will be stretched on a hot plate at 40° C using a mixture of 60% 2- butoxyethanol and 10% ethanol in water.
  • Sections will be covered by a plastic film, excess butoxyethanol-ethanol mixture removed and the slides will be left overnight to dry in a 40° C oven Sections will then be deplasticized in a solution of equal volumes of xylene-chloroform for 30 to 60 minutes Standard staining procedures for light microscopy will then be performed on the prepared sections.
  • Statistics Data will be presented as the mean + standard error of the mean (SD) of the independent experiments Statistical significance will be determined by one way analysis of variance (ANOVA) and Fisher's PLSD test (StatView 4.01 ; Brain Power, Inc ,
  • Porcine progenitor endothelial cells will be isolated from pig peripheral blood by the method of Asahara et al. (Isolation of Putative progenitor endothelial cells for angiogenesis. Science 275:964-967) Monoclonal anti-CD34 antibodies will be coupled to magnetic beads and incubated with the leukocyte fraction of pig whole blood. After incubation, bound cells will be eluted and cultured in M-199 containing 20% fetal bovine serum and bovine brain extract. (Clonetics, San Diego, CA). Cells will be characterized by fluorescent antibodies to CD45, CD34, CD31, Flk-1, Tie-2, and E- selectin.
  • purified porcine progenitor endothelial cells will be transfected with vascular endothelial growth factor (VEGF) using an adenovirus vector expressing the VEGF cDNA according to the methods of Rosengart et al. (Six-month assessment of a phase I trial of angiogenic gene therapy for the treatment of coronary artery disease using direct intramyocardial administration of an adenovirus vector expressing the VEGF121 cDNA. Ann. Surg. 230(4):466- 470 (1999), incorporated herein by reference).
  • VEGF vascular endothelial growth factor
  • the transfected purified porcine progenitor cells expressing VEGF will be infused into the porcine femoral artery model after balloon injury and stent implantation as described in Example 4 using a double-balloon chamber infusion catheter (Cordis Corp) which isolates the stented portion of the femoral artery. Restenosis will be compared in balloon angioplasty stent- treated pigs infused with VEGF-transfected porcine progenitor cells as compared with pigs infused with un-transfected porcine progenitor endothelial cells. Expression of VEGF in the re- infused porcine progenitor endothelial cells will result in a decreased incidence and severity of restenosis in the anti-CD34 coated stents.
  • Stent Preparation - stents will be made from 316L stainless steel and will be cleaned and passivated by first washing in an anionic detergent in an ultrasonic cleaner and then soaked in hot nitric acid with agitation, followed by a final deionized water rinse.
  • Derivatized stents will be prepared as follows - stents will be dipped into a 2% mixture of N-(2-aminoethyl-3-aminopropyl) trimethoxysilane in 95% ethanol for three minutes, removed, air dried at room temperature and then cured for 10 minutes at 110°C. Polyethylene glycol (PEG) Spacer Coupling - Derivatized stents will be placed in
  • Tethered Antibody - Antibodies to endothelial cells will be immobilized to the PEG functionalized stents in a one-step carbodiimide coupling reaction by immersing the stents into 150 ml of 0.1M MES buffer (pH 4.5) into which 1.0 mg of murine anti-CD34 IgGi antibody is dissolved and incubated at 25 °C for two hours. Stents will be removed from the solution and rinsed five times with 50 ml of phosphate buffered saline (pH 7.2) with 0.02% Tween 20.
  • Reagents include: N-(2-aminoethyl-3-aminopropyl)trimethoxysilane (Degussa-
  • Dicarboxymethyl-poly(ethylene glycol) [MW 3400] (Shearwater, Huntsville, AL).

Abstract

This invention provides compositions and methods for producing a medical device such as a stent or a synthetic graft, coated with a matrix and an antibody which reacts with an endothelial cell antigen. The matrix coating the medical device may be composed of synthetic material, such as polyurethane, poly-L-lactic acid, cellulose ester or polyethylene glycol. In another embodiment, the matrix is composed of naturally occurring materials, such as collagen, fibrin, elastin, amorphous carbon. In a third embodiment, the matrix may be composed of fullerenes which range from about C60 to about C100. The antibodies promote adherence of endothedial cells on the medical device. The antibodies may be mixed with the matrix or covalently tethered through a linker molecule to the matrix. Following adherence to the medical device, the endothelial cells differentiate and proliferate on the medical device. The antibodies may be different types of monoclonal antibodies.

Description

COATING THAT PROMOTES ENDOTHELIAL CELL ADHERENCE
This application claims the benefit of U S Provisional Application Nos 60/189,674 filed March 15, 2000 and 60/201,789 filed May 4, 2000
FIELD OF THE INVENTION
The present invention relates to the field of medical devices implanted in vessels within the body More particularly, the present invention relates to stents or synthetic grafts implanted in blood vessels that incorporate a matπx which promotes adherence of endothelial cells to the stent or synthetic graft
BACKGROUND OF THE INVENTION
Atherosclerosis is one of the leading causes of death and disability in the world Atherosclerosis involves the deposition of fatty plaques on the lumenal surface of arteries The deposition of fatty plaques on the lumenal surface of the artery causes narrowing of the cross- sectional area of the artery Ultimately, this deposition blocks blood flow distal to the lesion causing ischemic damage to the tissues supplied by the artery
Coronary artenes supply the heart with blood Coronary artery atherosclerosis disease (CAD) is the most common, serious, chronic, life-threatening illness in the United States, affecting more than 11 million persons The social and economic costs of coronary atherosclerosis vastly exceed that of most other diseases Narrowing of the coronary artery lumen causes destruction of heart muscle resulting first in angina, followed by myocardial infarction and finally death There are over 1 5 million myocardial infarctions in the United States each year Six hundred thousand (or 40%) of those patients suffer an acute myocardial infarction and more than three hundred thousand of those patients die before reaching the hospital (Harrison's Principles of Internal Medicine.14th Edition. 1998)
CAD can be treated using percutaneous translumenal coronary balloon angioplasty (PTCA) More than 400,000 PTCA procedures are performed each year in the United States In PTCA, a balloon catheter is inserted into a peripheral artery and threaded through the arterial system into the blocked coronary artery The balloon is then inflated, the artery stretched, and the obstructing fatty plaque flattened, thereby increasing the cross-sectional flow of blood through the affected artery The therapy, however, does not usually result in a permanent opening of the affected coronary artery As many as 50% of the patients who are treated by PTCA require a repeat procedure within six months to correct a re-narrowing of the coronary artery. Medically, this re-narrowing of the artery after treatment by PTCA is called restenosis. Acutely, restenosis involves recoil and shrinkage of the vessel. Subsequently, recoil and shrinkage of the vessel are followed by proliferation of medial smooth muscle cells in response to injury of the artery from PTCA In part, proliferation of smooth muscle cells is mediated by release of various inflammatory factors from the injured area including thromboxane A2> platelet derived growth factor (PDGF) and fibroblast growth factor (FGF). A number of different techniques have been used to overcome the problem of restenosis, including treatment of patients with various pharmacological agents or mechanically holding the artery open with a stent. (Harrison's Principles of Internal Medicine.14th Edition. 1998 h
Of the various procedures used to overcome restenosis, stents have proven to be the most effective Stents are metal scaffolds that are positioned in the diseased vessel segment to create a normal vessel lumen Placement of the stent in the affected arterial segment prevents recoil and subsequent closing of the artery. Stents can also prevent local dissection of the artery along the medial layer of the artery By maintaining a larger lumen than that created using PTCA alone, stents reduce restenosis by as much as 30%. Despite their success, stents have not eliminated restenosis entirely (Suryapranata et al 1998 Randomized comparison of coronary stenting with balloon angioplasty in selected patients with acute myocardial infarction. Circulation 97:2502-2502). Narrowing of the arteries can occur in vessels other than the coronary arteries, including the aortoiliac, infrainguinal, distal profunda femoris, distal popliteal, tibial, subclavian and mesenteric arteries. The prevalence of peripheral artery atherosclerosis disease (PAD) depends on the particular anatomic site affected as well as the criteria used for diagnosis of the occlusion. Traditionally, physicians have used the test of intermittent claudication to determine whether PAD is present. However, this measure may vastly underestimate the actual incidence of the disease in the population Rates of PAD appear to vary with age, with an increasing incidence of PAD in older individuals Data from the National Hospital Discharge Survey estimate that every year, 55,000 men and 44,000 women had a first-listed diagnosis of chronic PAD and 60,000 men and 50,000 women had a first-listed diagnosis of acute PAD. Ninety-one percent of the acute PAD cases involved the lower extremity. The prevalence of comorbid CAD in patients with PAD can exceed 50%. In addition, there is an increased prevalence of cerebrovascular disease among patients with PAD.
PAD can be treated using percutaneous translumenal balloon angioplasty (PTA). The use of stents in conjunction with PTA decreases the incidence of restenosis. However, the post-operative results obtained with medical devices such as stents do not match the results obtained using standard operative revascularization procedures, i.e., those using a venous or prosthetic bypass material. (Principles of Surgery. Schwartz et al. eds., Chapter 20, Arterial Disease, 7th Edition, McGraw-Hill Health Professions Division, New York 1999). Preferably, PAD is treated using bypass procedures where the blocked section of the artery is bypassed using a graft. (Principles of Surgery. Schwartz et al. eds., Chapter 20, Arterial Disease. 7th Edition, McGraw-Hill Health Professions Division, New York 1999). The graft can consist of an autologous venous segment such as the saphenous vein or a synthetic graft such as one made of polyester, polytetrafluoroethylene (PTFE), or expanded polytetrafluoroethylene (ePTFE). The post-operative patency rates depend on a number of different factors, including the lumenal dimensions of the bypass graft, the type of synthetic material used for the graft and the site of outflow. Restenosis and thrombosis, however, remain significant problems even with the use of bypass grafts. For example, the patency of infrainguinal bypass procedures at 3 years using an ePTFE bypass graft is 54% for a femoral-popliteal bypass and only 12% for a femoral-tibial bypass.
Consequently, there is a significant need to improve the performance of both stents and synthetic bypass grafts in order to further reduce the morbidity and mortality of CAD and PAD.
With stents, the approach has been to coat the stents with various anti-thrombotic or anti-restenotic agents in order to reduce thrombosis and restenosis. For example, impregnating stents with radioactive material appears to inhibit restenosis by inhibiting migration and proliferation of myofibroblasts. (U.S. Patent Nos. 5,059,166, 5,199,939 and 5,302, 168). Irradiation of the treated vessel can pose safety problems for the physician and the patient. In addition, irradiation does not permit uniform treatment of the affected vessel. Alternatively, stents have also been coated with chemical agents such as heparin or phosphorylcholine, both of which appear to decrease thrombosis and restenosis. Although heparin and phosphorylcholine appear to markedly reduce restenosis in animal models in the short term, treatment with these agents appears to have no long-term effect on preventing restenosis. Additionally, heparin can induce thrombocytopenia, leading to severe thromboembolic complications such as stroke. Nonetheless, it is not feasible to load stents with sufficient therapeutically effective quantities of either heparin or phosphorylcholine to make treatment of restenosis in this manner practical.
Synthetic grafts have been treated in a variety of ways to reduce postoperative restenosis and thrombosis. (Bos et al. 1998. Small-Diameter Vascular Graft Prostheses:Current Status Archives Physio. Biochem. 106: 100-115). For example, composites of polyurethane such as meshed polycarbonate urethane have been reported to reduce restenosis as compared with ePTFE grafts. The surface of the graft has also been modified using radiofrequency glow discharge to add polyterephalate to the ePTFE graft. Synthetic grafts have also been impregnated with biomolecules such as collagen. However, none of these approaches has significantly reduced the incidence of thrombosis or restenosis over an extended period of time.
Because endothelial cells possess certain intrinsic characteristics such as cell regulatory molecules that decrease the incidence of thrombosis or restenosis, stimulating the development of an endothelial cell monolayer on the surface of stents or synthetic grafts may prevent both restenosis and thrombosis. (Belle et al. 1997. Stent Endothelialization. Circulation 95:438-448; Bos et al. 1998. Small-Diameter Vascular Graft Prostheses: Current Status Archives Phvsio. Biochem. 106: 100-115).
Endothelial cells have been deposited on the surface of stents by local delivery of vascular endothelial growth factor (VEGF), an endothelial cell mitogen, after implantation of the stent. (Belle et al. 1997. Stent Endothelialization. Circulation 95:438-448.). Because the application of VEGF can have systemic as well as local effects, this form of treatment may be unreliable.
Synthetic grafts have also been seeded with endothelial cells, but the clinical results with endothelial seeding have been generally poor, i.e., low post-operative patency rates (Lio et al. 1998. New concepts and Materials in Microvascular Grafting: Prosthetic Graft Endothelial Cell Seeding and Gene Therapy. Microsurgery 18:263-256).
Accordingly, there is a need for development of new methods and compositions for coating medical devices, including stents and synthetic grafts, with endothelial cells. This type of coating will not only prevent restenosis, but also thromboembolic complications resulting from stent implantation. Methods and compositions that provide such improvement will eliminate the drawbacks of previous technology and have a significant positive impact on the morbidity and mortality associated with CAD and PAD. It is the object of this invention to prepare stents and synthetic grafts coated in such a manner as to stimulate adherence of endothelial cells to a medical device such as a stent or synthetic graft.
SUMMARY OF THE INVENTION
The invention provides methods and compositions for coating medical devices with a matrix that promotes adherence of endothelial cells to a medical device. The matrix incorporates antibodies that stimulate adherence of endothelial cells to the surface of the medical device.
As used herein, "medical device" refers to a device that is introduced temporarily or permanently into a mammal for the prophylaxis or therapy of a medical condition. These devices include any that are introduced subcutaneously, percutaneously or surgically to rest within an organ, tissue or lumen. Medical devices may include stents, covered stents such as those covered with polytetrafluoroethylene (PTFE), or expanded polytetrafluoroethylene (ePTFE), synthetic grafts, artificial heart valves, artificial hearts and fixtures to connect the prosthetic organ to the vascular circulation, venous valves, abdominal aortic aneurysm (AAA) grafts, inferior venal caval filters, permanent drug infusion catheters, embolic coils, embolic materials used in vascular embolization (e.g., PVA foams), and vascular sutures.
Coating of the medical device with the compositions and methods of this invention may stimulate the development of an endothelial cell layer on the surface of the medical device, thereby preventing restenosis as well as other thromboembolic complications that result from implantation of the medical device.
Synthetic grafts and stents can be used for treating CAD or PAD. A stent or synthetic graft may be coated with a matrix incorporating antibodies that stimulate adherence of circulating progenitor endothelial cells to the medical device. The antibodies may comprise monoclonal antibodies reactive with endothelial cell surface antigens such as CD34, an antigen expressed on the surface of progenitor endothelial cells. Fab fragments of the monoclonal antibody may be used. In another embodiment, monoclonal antibodies directed against other endothelial surface antigens such as KDR or Tie-2, may also be used. In one embodiment, a single type of antibody that reacts with one antigen may be used. Alternatively, a plurality of different antibodies directed against different endothelial cell surface antigens may be mixed together and added to the matrix.
The matrix coating the medical device may be composed of synthetic material, such as polyurethane, poly-L-lactic acid, cellulose ester or polyethylene glycol. In another embodiment, the matrix is composed of naturally occurring materials, such as collagen, fibrin, elastin or amorphous carbon. The matrix may comprise several layers with a first layer being composed of synthetic or naturally occurring materials and a second layer composed of antibodies. The layers may be ordered sequentially, with the first layer directly in contact with the stent or synthetic graft surface and the second layer having one surface in contact with the first layer and the opposite surface in contact with the vessel lumen. In a third embodiment, the matrix may comprise fullerenes, where the fullerenes range from about C60 to about CIOO. The fullerenes may also be arranged as nanotubes, that incorporate molecules or proteins. The fullerene matrix may also be mixed with PTFE or ePTFE, or antibodies. Alternatively, the PTFE or ePTFE may be layered first on the medical device followed by a second layer of fullerenes.
The matrix may be noncovalently or covalently attached to the medical device. Antibodies may be covalently attached to the matrix using hetero- or homobifunctional cross- linking reagents.
Methods of treatment of atherosclerosis are also provided. The artery may be the either a coronary artery or a peripheral artery such as the femoral artery.
Brief Description of the Figures
Figure 1 shows an antibody tethered covalently to the matrix by a cross-linking molecule Figure 2 shows a diagram of the C60O molecule anchoring the matnx DETAILED DESCRIPTION OF THE INVENTION
Overview
The present invention provides methods and compositions that involve coating a medical device such as a stent or synthetic graft with a matrix which is then used to coat the medical device In one embodiment, the matrix incorporates a therapeutically effective amount of at least one type of antibody that promotes adherence of endothelial cells to the medical device Following adherence, the endothelial cells differentiate and proliferate on the surface of the matrix The presence of endothelial cells on the medical device reduces the occurrence of restenosis and thrombosis after medical device implantation into a vessel
As used herein, the term "antibody" refers to one type of monoclonal or polyclonal antibody, where the monoclonal or polyclonal antibody binds to one antigen or a functional equivalent of that antigen The term antibody encompasses any fragment of an antibody such as Fab, F(ab')2 or Fc fragments (An antibody encompasses a plurality of individual antibody molecules equal to 6 022 x 1023 molecules per mole of antibody)
As used herein, a "therapeutically effective amount of the antibody" means the amount of an antibody that promotes adherence of endothelial cells to the medical device The amount of an antibody needed to practice the claimed invention varies with the nature of the antibody used For example, the amount of an antibody used will depend on the binding constant between the antibody and the antigen against which it reacts It is well known to those of ordinary skill in the art how to determine therapeutically effective amounts of an antibody to use with a particular antigen.
As used herein, "medical device" refers to a device that is introduced temporarily or permanently into a mammal for the prophylaxis or therapy of a medical condition These devices include any that are introduced subcutaneously, percutaneously or surgically to rest within an organ, tissue or lumen Medical devices may include, stents, covered stents such as those covered with PTFE, or ePTFE, synthetic grafts, artificial heart valves, artificial hearts and fixtures to connect the prosthetic organ to the vascular circulation, venous valves, abdominal aortic aneurysm (AAA) grafts, infenor venal caval filters, permanent drug infusion catheters, embolic coils, embolic materials used in vascular embolization (e g , PVA foams), and vascular sutures As used herein, "restenosis" refers to the accumulation of a layer of smooth muscle cells and matrix protein in the intima of an arterial wall Vessels may become obstructed because of restenosis. After PTCA or PTA, smooth muscle cells from the media and adventitia, which are not normally present in the intima, proliferate and migrate to the intima and secrete proteins, forming an accumulation of smooth muscle cells and matrix protein within the intima. This accumulation causes a narrowing of the lumen of the artery, reducing blood flow distal to the narrowing. As used herein, "inhibition of restenosis" refers to the inhibition of migration and proliferation of smooth muscle cells accompanied by prevention of protein secretion so as to prevent restenosis and the complications arising therefrom. The subjects that can be treated using the methods and compositions of this invention may be a mammal, or more specifically, a human, dog, cat, pig, rodent or monkey.
The methods of the present invention may be practiced in vivo or in vitro.
The term "endothelial cell" refers to endothelial cells at any developmental stage, from progenitor to mature. Fully differentiated endothelial cells may be isolated from an artery or vein such as a human umbilical vein, while progenitor endothelial cells are isolated from peripheral blood or bone marrow. The endothelial cells are bound to the medical devices by incubation of the endothelial cells with a medical device coated with the matrix that incorporates an antibody or other agent that adheres to endothelial cells.
The methods of this invention may be practiced on any artery or vein. Included within the scope of this invention is atherosclerosis of any artery including coronary, infrainguinal, aortoiliac, subclavian, mesenteric and renal arteries. Other types of vessel obstructions, such as those resulting from a dissecting aneurysm are also encompassed by the invention.
The medical device may be coated with endothelial cells after insertion into a vessel. Alternatively, the medical device is coated with the endothelial cells before insertion of the medical device. In either case, the presence of endothelial cells on the lumenal surface of the medical device inhibits or prevents restenosis and thrombosis.
Endothelial Cells
Human umbilical vein endothelial cells (HUVEC) are obtained from umbilical cords according to the methods of Jaffe, et al., J. Clin. Invest.. 52:2745-2757, 1973, incorporated herein by reference. Briefly, cells are stripped from the blood vessel walls by treatment with collagenase and cultured in gelatin-coated tissue culture flasks in Ml 99 medium containing 10% low endotoxin fetal calf serum, 90 ug/ml preservative-free porcine heparin, 20 ug/ml endothelial cell growth supplement (ECGS), glutamine and antibodies.
Progenitor endothelial cells are isolated from human peripheral blood according to the methods of Asahara et al. (Isolation of putative progenitor endothelial cells for angiogenesis. Science 275:964-967. 1997, incorporated herein by reference). Magnetic beads coated with antibody to CD34 are incubated with human peripheral blood. After incubation, bound cells are eluted and can be cultured in M- 199 containing 20% fetal bovine serum and bovine brain extract. (Clonetics, San Diego, CA). Cells are characterized by fluorescent antibodies to CD45, CD34, CD31, Flk-1, Tie-2, and E-selectin. Endothelial cells are transfected with any mammalian expression vectors that contains any cloned genes encoding proteins such as platelet derived growth factor (PDGF), fibroblast growth factor (FGF), or nitric oxide synthase (NOS) using conventional methods. (See, for example, mammalian expression vectors and transfection kits commercially available from Stratagene, San Diego, CA). For example, purified porcine progenitor endothelial cells are transfected with vascular endothelial growth factor (VEGF) using an adenoviral expression vector expressing the VEGF cDNA according to the methods of Rosengart et al. (Six-month assessment of a phase I trial of angiogenic gene therapy for the treatment of coronary artery disease using direct intramyocardial administration of an adenovirus vector expressing the VEGF121 cDNA. Ann. Surg. 230(4):466-470 (1999), incorporated herein by reference).
Antibodies
Monoclonal antibodies useful in the method of the invention may be produced according to the standard techniques of Kohler and Milstein (Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 265:495-497, 1975, incorporated herein by reference). Endothelial cells can be used as the immunogen to produce monoclonal antibodies directed against endothelial cell surface antigens.
Monoclonal antibodies directed against endothelial cells are prepared by injecting HUVEC or purified progenitor endothelial cells into a mouse or rat. After a sufficient time, the mouse is sacrificed and spleen cells are obtained. The spleen cells are immortalized by fusing them with myeloma cells or with lymphoma cells, generally in the presence of a non-ionic detergent, for example, polyethylene glycol. The resulting cells, which include the fused hybridomas, are allowed to grow in a selective medium, such as HAT-medium, and the surviving cells are grown in such medium using limiting dilution conditions. The cells are grown in a suitable container, e.g., microtiter wells, and the supernatant is screened for monoclonal antibodies having the desired specificity, i.e , reactivity with endothelial cell antigens
Various techniques exist for enhancing yields of monoclonal antibodies such as injection of the hybridoma cells into the peritoneal cavity of a mammalian host which accepts the cells and then harvesting the ascites fluid Where an insufficient amount of monoclonal antibody collects in the ascites fluid, the antibody is harvested from the blood of the host. Various conventional ways exist for isolation and purification of monoclonal antibodies so as to free the monoclonal antibodies from other proteins and other contaminants.
Also included within the scope of the invention are useful binding fragments of anti-endothelial cell monoclonal antibodies such as the Fab, F(ab')2, or Fc fragments of these monoclonal antibodies. The antibody fragments are obtained by conventional techniques. For example, useful binding fragments may be prepared by peptidase digestion of the antibody using papain or pepsin.
Antibodies of the invention are directed to an antibody of the IgG class from a murine source; however, this is not meant to be a limitation. The above antibody and those antibodies having functional equivalency with the above antibody, whether from a murine source, mammalian source including human, or other sources, or combinations thereof are included within the scope of this invention, as well as other classes such as IgM, IgA, IgE, and the like, including isotypes within such classes. In the case of antibodies, the term "functional equivalency" means that two different antibodies each bind to the same antigenic site on an antigen, in other words, the antibodies compete for binding to the same antigen. The antigen may be on the same or different molecule.
In one embodiment, monoclonal antibodies reacting with the endothelial cell surface antigen CD34 are used. Anti-CD34 monoclonal antibodies attached to a solid support have been shown to capture progenitor endothelial cells from human peripheral blood. After capture, these progenitor cells are capable of differentiating into endothelial cells. (Asahara et al. 1997. Isolation of putative progenitor endothelial cells for angiogenesis. Science 275:964-967.) Hybridomas producing monoclonal antibodies directed against CD34 can be obtained from the American Type Tissue Collection. (Rockville, MD). In another embodiment, monoclonal antibodies reactive with endothelial cell surface antigens Flk-1 or Tie-2 are used. Polyclonal antibodies reactive against endothelial cells isolated from the same species as the one receiving the medical device implant may also be used.
Stent The term "stent" herein means any medical device which when inserted into the lumen of a vessel expands the cross-sectional lumen of a vessel. The term "stent" includes covered stents such as those covered with PTFE or ePTFE. In one embodiment, this includes stents delivered percutaneously to treat coronary artery occlusions or to seal dissections or aneurysms of the splenic, carotid, iliac and popliteal vessels. In another embodiment, the stent is delivered into a venous vessel. The stent can be composed of polymeric or metallic structural elements onto which the matrix is applied or the stent can be a composite of the matrix intermixed with a polymer. For example, a deformable metal wire stent can be used, such as that disclosed in U.S. Pat. No. 4,886,062 to Wiktor, incorporated herein by reference. A self-expanding stent of resilient polymeric material such as that disclosed in published international patent application WO91/12779 "Intraluminal Drug Eluting Prosthesis", incorporated herein by reference, can also be used. Stents may also be manufactured using stainless steel, polymers, nickel-titanium, tantalum, gold, platinum-iridium, or Elgiloy and MP35N and other ferrous materials. Stents are delivered through the body lumen on a catheter to the treatment site where the stent is released from the catheter, allowing the stent to expand into direct contact with the lumenal wall of the vessel. It will be apparent to those skilled in the art that other self-expanding stent designs (such as resilient metal stent designs) could be used with the antibodies and matrices of this invention.
Synthetic Graft The term "synthetic graft" means any artificial prosthesis having biocompatible characteristics. In one embodiment this includes synthetic grafts made of Dacron (polyethylene terephthalate, PET) or Teflon (ePTFE). In another embodiment, synthetic grafts are composed of polyurethane. In yet a third embodiment, a synthetic graft is composed of an inner layer of meshed polycarbonate urethane and an outer layer of meshed Dacron. It will be apparent to those skilled in the art that any biocompatible synthetic graft can be used with the antibodies and matrices of this invention. (Bos et al. 1998. Small-Diameter Vascular Prostheses: Current Status. Archives Physio Biochem. 106:100-115, incorporated herein by reference). Synthetic grafts can be used for end-to-end anastomosis of vessels or for bypass of a diseased vessel segment.
Matrix
(A) Synthetic Materials - The matrix that is used to coat the stent or synthetic graft may be selected from synthetic materials such as polyurethane, segmented polyurethane-urea/heparin, poly-L-lactic acid, cellulose ester or polyethylene glycol. (B) Naturally Occurring Material - The matrix may be selected from naturally occurring substances such as collagen, laminin, heparin, fibrin, cellulose or carbon. A primary requirement for the matrix is that it be sufficiently elastic and flexible to remain unruptured on the exposed surfaces of the stent or synthetic graft. (C) Fullerenes - The matrix may also comprise a fullerene (the term "fullerene" encompasses a plurality of fullerene molecules). Fullerenes are carbon-cage molecules. The number of carbon
(C) molecules in a fullerene species varies from about C60 to about CIOO. Fullerenes are produced by high temperature reactions of elemental carbon or of carbon-containing species by processes well known to those skilled in the art; for example, by laser vaporization of carbon, heating carbon in an electric arc or burning of hydrocarbons in sooting flames. (U.S. Patent No. 5,292,813, to Patel et al., incorporated herein by reference; U.S. Patent No. 5,558,903 to Bhushan et al., incorporated herein by reference). In each case, a carbonaceous deposit or soot is produced. From this soot, various fullerenes are obtained by extraction with appropriate solvents, such as toluene. The fullerenes are separated by known methods, in particular by high performance liquid chromatography (HPLC). Fullerenes may be synthesized or obtained commercially from Dynamic Enterprises, Ltd., Berkshire, England or Southern Chemical Group, LLC, Tucker, Georgia.
Fullerenes may be deposited on surfaces in a variety of different ways, including, sublimation, laser vaporization, sputtering, ion beam, spray coating, dip coating, roll-on or brush coating as disclosed in U.S. Patent No. 5,558,903.
An important feature of fullerenes is their ability to form "activated carbon." The fullerene electronic structure is a system of overlapping pi-orbitals, such that a multitude of bonding electrons are cooperatively presented around the surface of the molecule. (Chemical and Engineering News. Apr. 8, 1991, page 59, incorporated herein by reference). As forms of activated carbon, fullerenes exhibit substantial van der Waals forces for weak interactions. The adsorptive nature of the fullerene surface may lend itself to additional modifications for the purpose of directing specific cell membrane interactions. For example, specific molecules that possess chemical properties that selectively bind to cell membranes of particular cell types or to particular components of cell membranes, e.g., lectins or antibodies, can be adsorbed to the fullerene surface. The fullerene surface may also be chemically modified to present specifically reactive groups to the cell membrane, e.g., oxidants or reductants. Attachment of different molecules to the fullerene surface may be manipulated to create surfaces that selectively bind various cell types, e.g., epithelial cells, fibroblasts, primary explants, or T-cell subpopulations. U.S. Patent No. 5,310,669 to Richmond et al., incorporated herein by reference; Stephen R. Wilson, Biological Aspects of Fullerenes, Fullerenes: Chemistry. Physics and Technology. Kadish et al. eds., John Wiley & Sons, NY 2000, incorporated herein by reference.
Fullerenes may also form nanotubes that incorporate other atoms or molecules. (Liu et al. Science 280: 1253-1256 (1998), incorporated herein by reference). The synthesis and preparation of carbon nanotubes is well known in the art. (U.S. Patent No. 5,753,088 to Oik et al., and U.S. Patent No. 5,641,466 to Ebbsen et al., both incorporated herein by reference). Molecules such as proteins may also be incorporated inside carbon nanotubes. For example, nanotubes may be filled with the enzymes, e.g., Zn2Cd -metallothionein, cytochromes C and C3, and beta-lactamase after cutting the ends of the nanotube. (Davis et al. Inorganica Chim. Acta 272:261 (1998); Cook et al. Full Sci. Tech. 5(4):695 (1997), both incorporated herein by reference).
Three dimensional fullerene structures may also be used. U.S. Patent No. 5,338,571 to Mirkin et al., incorporated herein by reference, discloses three-dimensional, multilayer fullerene structures that are formed on a substrate surface by (i) chemically modifying fullerenes to provide a bond-forming species; (ii) chemically treating a surface of the substrate to provide a bond-forming species effective to covalently bond with the bond-forming species of the fullerenes in solution; and, (iii) contacting a solution of modified fullerenes with the treated substrate surface to form a fullerene layer covalently bonded to the treated substrate surface.
(D) Application of the Matrix to the Medical Device
The matrix should adhere tightly to the surface of the stent or synthetic graft. Preferably, this is accomplished by applying the matrix in successive thin layers. Each layer of matrix may incorporate the antibodies. Alternatively, antibodies may be applied only to the layer in direct contact with the vessel lumen. Different types of matrices may be applied successively in succeeding layers. The antibodies may be covalently or noncovalently coated on the matrix after application of the matrix to the stent.
In order to coat a medical device such as a stent, the stent is dipped or sprayed with a liquid solution of the matrix of moderate viscosity. After each layer is applied, the stent is dried before application of the next layer. In one embodiment, a thin, paint-like matrix coating does not exceed an overall thickness of 100 microns.
For example, a suitable matrix coating solution is prepared by dissolving 480 milligrams (mg) of a drug carrier, such as poly-D, L-lactid (available as R203 of Boehringer Inc., Ingelheim, Germany) in 3 milliliters (ml) of chloroform under aseptic conditions. In principle, however, any biodegradable (or non-biodegradable) matrix that is blood-and tissue-compatible (biocompatible) and can be dissolved, dispersed or emulsified may be used as the matrix if, after application, it undergoes relatively rapid drying to a self-adhesive lacquer- or paint-like coating on the medical device. For example, coating a stent with fibrin is well known to one of ordinary skill in the art. In U.S. Patent No. 4,548,736 issued to Muller et al., incorporated herein by reference, fibrin is clotted by contacting fibrinogen with thrombin. Preferably, the fibrin in the fibrin- containing stent of the present invention has Factor XIII and calcium present during clotting, as described in U.S. Pat. No. 3,523,807 issued to Gerendas, incorporated herein by reference, or as described in published European Patent Application 0366564, incorporated herein by reference, in order to improve the mechanical properties and biostability of the implanted device. Preferably, the fibrinogen and thrombin used to make fibrin in the present invention are from the same animal or human species as that in which the stent will be implanted in order to avoid any inter-species immune reactions, e g., human anti-cow. The fibrin product can be in the form of a fine, fibrin film produced by casting the combined fibrinogen and thrombin in a film and then removing moisture from the film osmotically through a semipermeable membrane. In the European Patent Application 0366564, a substrate (preferably having high porosity or high affinity for either thrombin or fibrinogen) is contacted with a fibrinogen solution and with a thrombin solution. The result is a fibrin layer formed by polymerization of fibrinogen on the surface of the medical device. Multiple layers of fibrin applied by this method could provide a fibrin layer of any desired thickness. Alternatively, the fibrin can first be clotted and then ground into a powder which is mixed with water and stamped into a desired shape in a heated mold (U.S. Patent No. 3,523,807). Increased stability can also be achieved in the shaped fibrin by contacting the fibrin with a fixing agent such as glutaraldehyde or formaldehyde. These and other methods known by those skilled in the art for making and forming fibrin may be used in the present invention.
If a synthetic graft is coated with collagen, the methods for preparing collagen and forming it on synthetic graft devices are well known as set forth in U.S. Patent No. 5,851,230 to Weadock et al., incorporated herein by reference. This patent describes methods for coating a synthetic graft with collagen. Methods for adhering collagen to a porous graft substrate typically include applying a collagen dispersion to the substrate, allowing it to dry and repeating the process. Collagen dispersions are typically made by blending insoluble collagen (approximately 1- 2% by weight) in a dispersion at acidic pH (a pH in a range of 2 to 4). The dispersion is typically injected via syringe into the lumen of a graft and massaged manually to cover the entire inner surface area with the collagen slurry. Excess collagen slurry is removed through one of the open ends of the graft Coating and drying steps are repeated several times to provide sufficient treatment
In yet another embodiment, the stent or synthetic graft is coated with amorphous carbon In U S Patent No 5,198,263, incorporated herein by reference, a method for producing a high-rate, low-temperature deposition of amorphous carbon films in the presence of a fluoπnated or other halide gas is descnbed Deposition according to the methods of this invention can be performed at less than 100° C, including ambient room temperature, with a radio- frequency, plasma-assisted, chemical-vapor deposition process The amorphous carbon film produced using the methods of this invention adheres well to many types of substrates, including for example glasses, metals, semiconductors, and plastics
Attachment of a fullerene moiety to reactive ammo group sites of an amine- containing polymer to form the fullerene-graft, amine-containing polymers may be performed as described in U S Patent No 5,292,813 Chemical modification in this manner allows for direct incorporation of the fullerenes into the stent In another embodiment, the fullerenes may be deposited on the surface of the stent or synthetic grafts as described above (see, WO 99/32184 to Leone et al , incorporated by reference) Fullerenes may also be attached through an aldehyde bond (Yamago et al , Chemical Denvatization of Organofullerenes through Oxidation, Reduction and C-0 and C-C Bond Forming Reactions J Org Chem . 58 4796-4798 (1998), incorporated herein by reference) C60O may also be attached directly through an epoxide group on the fullerene to a stent The attachment is through a covalent linkage to the oxygen This compound and the protocols for coupling are commercially available from BuckyUSA (BuckyUSA, Houston, Texas)
(E) Addition of Antibodies to the Matπx - Antibodies that promote adherence of progenitor endothelial cells can be incorporated into the matrix, either covalently or noncovalently Antibodies may be incorporated into each layer of matnx by mixing the antibodies with the matrix coating solution Alternatively, the antibodies may be covalently or noncovalently coated on to last layer of matrix that is applied to the medical device
In one embodiment, the antibodies are added to a solution containing the matπx For example, Fab fragments on anti-CD34 monoclonal antibody are incubated with a solution containing human fibrinogen at a concentration of between 500 and 800 mg/dl It will be appreciated that the concentration of anti-CD34 Fab fragment will vary and that one of ordinary skill in the art could determine the optimal concentration without undue experimentation The stent is added to the Fab/fibrin mixture and the fibπn activated by addition of concentrated thrombin (at a concentration of at least lOOOU/ml) The resulting polymerized fibrin mixture containing the Fab fragments incorporated directly into the matrix is pressed into a thin film (less than 0.12 cm) on the surface of the stent or synthetic graft. Virtually any type of antibody or antibody fragment can be incorporated in this manner into a matrix solution prior to coating of a stent or synthetic graft. In another embodiment, antibodies are covalently coupled to the matrix. In one embodiment, the antibodies are tethered covalently to the matrix through the use of hetero- or homobifunctional linker molecules. As used herein the term "tethered" refers to a covalent coupling of the antibody to the matrix by a linker molecule. The use of linker molecules in connection with the present invention typically involves covalently coupling the linker molecules to the matrix after it is adhered to the stent. After covalent coupling to the matrix, the linker molecules provide the matrix with a number of functionally active groups that can be used to covalently couple one or more types of antibody. Figure 1 provides an illustration of coupling via a cross-linking molecule. An endothelial cell, 1.01, binds to an antibody, 1.03, by a cell surface antigen, 1.02. The antibody is tethered to the matrix, 1.05-1.06, by a cross-linking molecule, 1.04. The matrix, 1.05-1.06, adheres to the stent, 1.07. The linker molecules may be coupled to the matrix directly (i.e., through the carboxyl groups), or through well-known coupling chemistries, such as, esterification, amidation, and acylation. The linker molecule may be a di- or tri-amine functional compound that is coupled to the matrix through the direct formation of amide bonds, and provides amine-functional groups that are available for reaction with the antibodies. For example, the linker molecule could be a polyamine functional polymer such as polyethyleneimine (PEI), polyallylamine (PALLA) or polyethyleneglycol (PEG). A variety of PEG derivatives, e.g., mPEG-succinimidyl propionate or mPEG-N-hydroxysuccinimide, together with protocols for covalent coupling, are commercially available from Shearwater Corporation, Birmingham, Alabama. (See also, Weiner et al., Influence of a poly-ethyleneglycol spacer on antigen capture by immobilized antibodies. J. Biochem. Biophys. Methods 45:211-219 (2000), incorporated herein by reference). It will be appreciated that the selection of the particular coupling agent may depend on the type of antibody used and that such selection may be made without undue experimentation. Mixtures of these polymers can also be used. These molecules contain a plurality of pendant amine-functional groups that can be used to surface-immobilize one or more antibodies.
Antibodies may be attached to C60O fullerene layers that have been deposited directly on the surface of the stent. Cross linking agents may be covalently attached to the fullerenes. The antibodies are then attached to the cross-linking agent, which in turn is attached to the stent. Figure 2 provides an illustration of coupling by C60O. The endothelial cell, 2.01, is bound via a cell surface antigen, 2 02, to an antibody, 2 03, which in turn is bound, covalently or non-covalently to the matrix, 2 04 The matrix, 2 04, is coupled covalently via C60O, 2 05, to the stent, 2 06
EXPERIMENTAL EXAMPLES
This invention is illustrated in the experimental details section which follows These sections set forth below the understanding of the invention, but are not intended to, and should not be construed to, limit in any way the invention as set forth in the claims which follow thereafter EXAMPLE 1
ADHERENCE OF HUMAN ENDOTHELIAL CELLS TO CD34 Fab-COATED STENTS
Materials and Methods 1 Cells HUVEC will be prepared from human umbilical cords by the method of Jaffe
(Jaffe, E A In "Biology of Endothelial Cells". E A Jaffe, ed , Martinus-Nijhoff, The Hague (1984), incorporated herein by reference) and cultured in Medium 199 supplemented with 20% fetal calf serum (FCS), L-glutamine, antibiotics, 130 ug/ml heparin and 1 2 mg/ml endothelial cell growth supplement (Sigma- Aldrich, St Louis, MO) Progenitor endothelial cells will be isolated from human peπpheral blood by the method of Asahara et al (Isolation of Putative progenitor endothelial cells for angiogenesis Science 275 964-967) Monoclonal anti-CD34 antibodies will be coupled to magnetic beads and incubated with the leukocyte fraction of human whole blood After incubation, bound cells will be eluted and cultured in M-199 containing 20% fetal bovine serum and bovine brain extract (Clonetics, San Diego, CA) Cells will be characterized by fluorescent antibodies to CD45, CD34, CD31, Flk-1, Tie-2, and E- selectin
2. Coating of Stents
A. R stents produced by Orbus International B.V. (Leusden, The Netherlands) will be incubated with human fibrinogen (Sigma, St. Louis, MO) 500-800 mg/ml together with Fab fragments of anti-CD34 monoclonal antibody and the fibrinogen will be polymerized by the addition of 1000 units/ml of thrombin. After incubation of the stent with the polymerized fibrin mixture containing the anti-CD34 monoclonal Fab fragments, the fibrin will be compressed into a thin film (less than 0.012 cm) against the R Stent. The R-Stent having the thin, fibrin film containing the Fab fragments will be washed three times with phosphate-buffered saline (PBS) containing 0.5% bovine serum albumin (BSA) at room temperature.
B. Alternatively, R stents will be coated with mPEG-succinimidyl propionate (Shearwater Corporation, Birmingham, Alabama). The succinimidyl group will be reacted with the anti-CD34 monoclonal Fab fragments (Fab-PEG coated R stents) according to the manufacturer's instructions to form a stable amide linkage between the PEG derivative and the Fab fragment.
3. Endothelial Cell binding assay
The fibrin-anti-CD34 Fab coated R-stents or Fab-PEG coated R stents will be incubated with isolated HUVEC or isolated progenitor endothelial cells at cellular concentrations of between 100,000 and 1 ,000,000 cells/ml in M199 containing 0.5% BSA at 37°C in a 5% CO2 humidified atmosphere. Prior to incubation with the stent, the HUVEC or progenitor endothelial cells will be labeled with [3H]-thymidine for 24 hours. After incubation of the labeled endothelial cells with the stents coated with fibrin and Fab anti-CD34 for between 4 and 72 hours, the stents will be removed from the solution and washed 5 times with Ml 99 containing 0.5% BSA. Bound endothelial cells will be removed by trypsinization and binding of labeled endothelial cells to the stents will be assessed by scintillation counting of [3H]-thymidine. As negative control, stents coated with fibrin alone or uncoated stents will be incubated with [3H]-thymidine-labeled endothelial cells. Results will be evaluated statistically using a t-test to determine differential binding. Stents coated with fibrin which incorporate monoclonal anti-CD34 Fab fragments will show significantly increased binding of endothelial cells as compared with uncoated stents.
EXAMPLE 2
PROLIFERATION OF HUMAN ENDOTHELIAL CELLS ON CD34 FAB-COATED STENTS Endothelial Cell Proliferation Assay
R stents coated with fibrin that incorporates anti-CD34 Fab fragments will be incubated with HUVEC or progenitor endothelial cells for between 4 and 72 hours in Ml 99 containing 0.5% BSA. After incubation of the stents with the HUVEC or progenitor endothelial cells, the stents will be washed 5 times with Ml 99 containing 0.5% BSA and then incubated with [3H]-thymidine. [3H]-thymidine incorporation will be assessed in washed and harvested HUVEC or progenitor endothelial cells (cells will be harvested with trypsin). Proliferation of HUVEC or progenitor endothelial cells on fibrin-coated stents will be compared with endothelial cell proliferation in standard microtiter dishes. Proliferation of HUVEC or progenitor endothelial cells on fibrin-coated stents will be equal to or greater than proliferation of endothelial cells in microtiter dishes
EXAMPLE 3
PRODUCTION OF MONOCLONAL ANTIBODIES REACTIVE WITH HUVEC AND PROGENITOR ENDOTHELIAL CELLS
BALB/c mice will be immunized, intraperitoneally 3-4 times at 2-4 weekly intervals, with 1.5 x 106 HUVEC in PBS or 1.5 x 106 progenitor endothelial cells and challenged 3 days prior to spleen-cell removal with 1.5 x 106 HUVEC or 1.5 x 106 progenitor endothelial cells. A spleen-cell suspension will be prepared, fused with the myeloma NS 1/1 AG4.1 and hybridomas grown up and cloned. To improve hybridoma growth and cloning efficiencies, 10% endothelial-cell conditioned medium (HUVEC) will be included in culture media. Initially, hybridoma culture supernatants will be tested for reactivity with HUVEC or progenitor endothelial cells by immunofluorescence flow cytometry (FACS). Briefly, HUVEC (1.5 x 104) or progenitor endothelial cells (1.5 x 104) will be incubated (30 min, 4°C) with undiluted hybridoma supernatant, washed and incubated with fluorescein-isothiocyanate (FITC)-sheep F(ab')2 anti- mouse Ig(100 ug/ml). Following final washing, the endothelial cells will be examined for monoclonal antibody binding by FACS analysis. Positive hybridoma supernatants will be screened on the human melanoma cell line MM- 170 to eliminate non-endothelial specific mAbs. Endothelial specificity will be further confirmed by screening of monoclonal antibodies on a panel of human tumor cell lines as well as human lymphocytes, monocytes, neutrophils, red cells and platelets. EXAMPLE 4
PORCINE BALLOON INJURY STUDIES
Implantation of antibody-covered stents will be performed in juvenile Yorkshire pigs weighing between 25 and 30 kg. Animal care will comply with the "Guide for the Care and Use of Laboratory Animals" (NIH publication No. 80-23, revised 1985). After an overnight fast, animals will be sedated with ketamine hydrochloride (20mg/kg). Following the induction of anesthesia with thiopental (12 mg kg) the animals will be intubated and connected to a ventilator that will administer a mixture of oxygen and nitrous oxide (1 :2 [vol/vol]). Anesthesia will be maintained with 0.5-2.5 vol% isoflurane. Antibiotic prophylaxis will be provided by an intramuscular injection of 1,000 mg of a mixture of procaine penicillin-G and benzathine penicillin-G (streptomycin).
Under sterile conditions, an arteriotomy of the left carotid artery will be performed and a 9F- introducer sheath will be placed in the left carotid artery. All animals will be given 7,500 IU of heparin sodium and 100 mg of acetylsalicylic acid intravenously. Additional 2,500 IU boluses of heparin will be administered periodically throughout the procedure in order to maintain an activated clotting time above 300 seconds. An 8F guiding catheter will be introduced through the carotid sheath and passed to the origin of the iliac artery. Angiography will be performed after the administration of lmg of isosorbide dinitrate and images will be analyzed using a quantitative coronary angiography system. A 3F-embolectomy catheter will be inserted into the common femoral artery, and passed distal to the segment selected for stent implantation. The embolectomy balloon will be inflated to a size 0.5 mm larger than the arterial segment and withdrawn twice to denude the vessel. Immediately after denudation, a fibrin-coated stent incorporating an Fab fragment of a monoclonal antibody will be inserted through the guiding catheter and deployed in the denuded segment of the femoral artery. Animals will be sacrificed both at 3 days and at eight weeks after stent implantation. The animal will first be sedated and anesthetized as described above. The stented femoral segments will be explanted and then placed in 4% paraformaldehyde in 0.1 M phosphate buffer pH 7.2 at 4° C for 48 h. A rectangular section of the vessel wall will be removed for further processing for electron microscopic evaluation of surface coverage of endothelial cells. This portion of the stented vessel will be placed in 0.15 cacodylate buffer and further fixed with 2.5% glutaraldehyde in 0.15 M cacodylate. The tissue will then be post-fixed with 0.1 M cacodylate buffer containing 1% OsO and 50 mM ferricyaπide (K3[Fe(CN)6]), and further processed. (Reduction in thrombotic events with heparin-coated Palmaz-Schatz stents in normal porcine coronary arteries, Circulation 93 423-430, incorporated herein by reference).
The remaining sections of the stented arterial segments will be impregnated with three changes of methy methacrylate as described by van Beusekom et al (Cardiovasc Pathol 5:69-76 (1996), incorporated herein by reference) Embedded arterial segments with the stent in place will be cut into sections 3 to 5 μm thick on a motor-driven rotary microtome (HM-350, Microm GmbH, Munich, Germany) using stainless steel disposable knives On chrome aluminum coated slides, sections will be stretched on a hot plate at 40° C using a mixture of 60% 2- butoxyethanol and 10% ethanol in water. Sections will be covered by a plastic film, excess butoxyethanol-ethanol mixture removed and the slides will be left overnight to dry in a 40° C oven Sections will then be deplasticized in a solution of equal volumes of xylene-chloroform for 30 to 60 minutes Standard staining procedures for light microscopy will then be performed on the prepared sections. Statistics: Data will be presented as the mean + standard error of the mean (SD) of the independent experiments Statistical significance will be determined by one way analysis of variance (ANOVA) and Fisher's PLSD test (StatView 4.01 ; Brain Power, Inc ,
Calabasas, Calif)- For data of treated and untreated segments of femoral arteries, a paired t test (StatView 4.01) will be used. A p value of <0.05 will be considered a statistically significant difference between the means. Animals treated with a stent incorporating an anti-porcine endothelial cell monoclonal Fab fragment will show increased endothelial cell coverage and significantly reduced restenosis as compared with controls having an uncoated stent implanted. EXAMPLE 5
TRANSFECTION OF PORCINE PROGENITOR ENDOTHELIAL CELLS
Porcine progenitor endothelial cells will be isolated from pig peripheral blood by the method of Asahara et al. (Isolation of Putative progenitor endothelial cells for angiogenesis. Science 275:964-967) Monoclonal anti-CD34 antibodies will be coupled to magnetic beads and incubated with the leukocyte fraction of pig whole blood. After incubation, bound cells will be eluted and cultured in M-199 containing 20% fetal bovine serum and bovine brain extract. (Clonetics, San Diego, CA). Cells will be characterized by fluorescent antibodies to CD45, CD34, CD31, Flk-1, Tie-2, and E- selectin. For example, purified porcine progenitor endothelial cells will be transfected with vascular endothelial growth factor (VEGF) using an adenovirus vector expressing the VEGF cDNA according to the methods of Rosengart et al. (Six-month assessment of a phase I trial of angiogenic gene therapy for the treatment of coronary artery disease using direct intramyocardial administration of an adenovirus vector expressing the VEGF121 cDNA. Ann. Surg. 230(4):466- 470 (1999), incorporated herein by reference).
The transfected purified porcine progenitor cells expressing VEGF will be infused into the porcine femoral artery model after balloon injury and stent implantation as described in Example 4 using a double-balloon chamber infusion catheter (Cordis Corp) which isolates the stented portion of the femoral artery. Restenosis will be compared in balloon angioplasty stent- treated pigs infused with VEGF-transfected porcine progenitor cells as compared with pigs infused with un-transfected porcine progenitor endothelial cells. Expression of VEGF in the re- infused porcine progenitor endothelial cells will result in a decreased incidence and severity of restenosis in the anti-CD34 coated stents.
EXAMPLE 6
PREPARATION OF AMINOSILANE PEO TETHERED ANTIBODIES
Stent Preparation - stents will be made from 316L stainless steel and will be cleaned and passivated by first washing in an anionic detergent in an ultrasonic cleaner and then soaked in hot nitric acid with agitation, followed by a final deionized water rinse.
Derivatized stents will be prepared as follows - stents will be dipped into a 2% mixture of N-(2-aminoethyl-3-aminopropyl) trimethoxysilane in 95% ethanol for three minutes, removed, air dried at room temperature and then cured for 10 minutes at 110°C. Polyethylene glycol (PEG) Spacer Coupling - Derivatized stents will be placed in
100ml of 0.1M MES buffer containing 10mM Dicarboxymethyl-PEG and 500 mg of EDC added and incubated at 25°C with constant stirring for two hours.
Tethered Antibody - Antibodies to endothelial cells will be immobilized to the PEG functionalized stents in a one-step carbodiimide coupling reaction by immersing the stents into 150 ml of 0.1M MES buffer (pH 4.5) into which 1.0 mg of murine anti-CD34 IgGi antibody is dissolved and incubated at 25 °C for two hours. Stents will be removed from the solution and rinsed five times with 50 ml of phosphate buffered saline (pH 7.2) with 0.02% Tween 20.
Reagents include: N-(2-aminoethyl-3-aminopropyl)trimethoxysilane (Degussa-
Huls); MES buffer - morpholine ethane sulfonic acid buffer (Sigma, St. Louis, MO); EDC - 1- ethyl-3-(3-dimethylaminopropyl) carbodiimide (Sigma, St. Louis, MO); Dicarboxymethyl-PEG -
Dicarboxymethyl-poly(ethylene glycol) [MW 3400] (Shearwater, Huntsville, AL).
Having described several different embodiments of the invention, it is not intended that the invention is limited to these embodiments it is intended that modifications and variations may be made by one skilled in the art without departing from the spirit and scope of the invention as defined in the claims.

Claims

We claim:
1. A medical device coated with a therapeutically effective amount of at least one type of antibody which reacts with an endothelial cell antigen and at least one layer of a matrix, wherein the matrix comprises a fullerene ranging from about C60 to about CIOO. 2. The medical device of claim 1, wherein the antibody is tethered covalently by a linker molecule to the last layer of the matrix coating the medical device.
3. The medical device of claim 2, wherein the fullerene is C60O.
4. The medical device of claim 1, wherein the antibody is a monoclonal antibody.
5. The medical device of claim 1, wherein the medical device is a stent. 6. The medical device of claim 1, wherein the medical device is a synthetic graft.
7. The medical device of claim 1, wherein the endothelial cell is a human cell.
8. The medical device of claim 4, wherein the monoclonal antibody reacts with endothelial cell surface antigen CD34.
9. The medical device of claim 4 or 8, wherein the monoclonal antibody comprises Fab or F(ab')2 fragments.
10. A medical device coated with a therapeutically effective amount of at least one type of antibody which reacts with an endothelial cell antigen and at least one layer of a matrix, wherein the matrix comprises polyurethane, segmented polyurethane-urea/heparin, poly-L-lactic acid, cellulose ester, polyethylene glycol, collagen, laminin, heparin, fibrin, cellulose or carbon. 1 1. The medical device of claim 10, wherein the antibody is tethered covalently by a linker molecule to the last layer of the matrix coating the medical device.
12. The medical device of claim 10, wherein the antibody is a monoclonal antibody.
13. The medical device of claim 10, wherein the medical device is a stent.
14. The medical device of claim 10, wherein the medical device is a synthetic graft. 15. The medical device of claim 10, wherein the endothelial cell is a human cell.
16. The medical device of claim 12, wherein the monoclonal antibody reacts with endothelial cell surface antigen, CD34.
17. The medical device of claim 12 or 16, wherein the monoclonal antibody comprises Fab or F(ab')2 fragments. 18. A composition for coating to a medical device comprising a matrix and a therapeutically effective amount of at least one type of antibody that reacts with an endothelial cell antigen. 19. The composition of claim 18, wherein the matrix comprises polyurethane, segmented polyurethane-urea/heparin, poly-L-lactic acid, cellulose ester, polyethylene glycol, collagen, laminin, heparin, fibrin, cellulose or carbon. 20 The composition of claim 18, wherein the matrix comprises a fullerene ranging from about C60 to about CIOO
21 The composition of claim 19 or 20, wherein the antibody is a monoclonal antibody
22 The composition of claim 21, wherein the endothelial cell is a human cell 23 The composition of claim 21, wherein the monoclonal antibody reacts with endothelial cell surface antigen, CD34
24 The composition of claim 22, wherein the monoclonal antibody compπses Fab or F(ab')2 fragments
25 A method for coating a medical device comprising the steps of (a) coating a medical device with at least one layer of a matrix comprising polyurethane, segmented polyurethane-urea hepaπn, poly-L-lactic acid, cellulose ester, polyethylene glycol, collagen, laminin, heparin, fibrin, cellulose, fullerene or carbon, and
(b) adding a therapeutically effective amount of at least one type of antibody which reacts with an endothelial cell antigen to the matrix coating the medical device 26 The method of claim 25, wherein the antibody is noncovalently coated on the last layer of the matrix coating the medical device
27 The method of claim 25, wherein the antibody is tethered covalently by a linker molecule to the last layer of the matrix coating the medical device
28 The method of claim 25, wherein the fullerene is C60O 29 A method of treating mammals for atherosclerosis comprising insertion of a medical device into an artery wherein the medical device is coated with a therapeutically effective amount of at least one type of antibody which reacts with an endothelial cell antigen and a matrix comprising a fullerene ranging from about C60 to CIOO 30 The method of treatment of claim 29, wherein the antibody is a monoclonal antibody 31 The method of treatment of claim 29, wherein the atherosclerosis is coronary artery atherosclerosis
32 The method of treatment of claim 30, wherein the monoclonal antibody comprises Fab or F(ab')2 fragments
33 A method for treating mammals for atherosclerosis comprising insertion into an artery of a medical device, wherein the medical device is coated with at least one layer of a matrix comprising polyurethane, segmented polyurethane-urea/heparin, poly-L-lactic acid, cellulose ester, polyethylene glycol, collagen, laminin, hepaπn, fibrin, cellulose or carbon and a therapeutically effective amount of at least one type of antibody which reacts with an endothelial cell antigen
34. The method of treatment of claim 33, wherein the antibody is a monoclonal antibody.
35. The method of treatment of claim 34, wherein the monoclonal antibody comprises Fab or F(ab')2 fragments.
36. A method for treating mammals for obstruction of a vessel comprising insertion into a vessel of a medical device coated with at least one layer of a matrix comprising polyurethane, segmented polyurethane-urea/heparin, poly-L-lactic acid, cellulose ester, polyethylene glycol, collagen, laminin, heparin, fibrin, cellulose or carbon and a therapeutically effective amount of at least one type of antibody which reacts with an endothelial cell antigen.
37. The method of treatment of claim 36, wherein the antibody is a monoclonal antibody. 38. A method for treating mammals for obstruction of a vessel comprising insertion into a vessel of a medical device coated with at least one layer of a matrix comprising a fullerene ranging from about C60 to CIOO and a therapeutically effective amount of at least one type of antibody which reacts with an endothelial cell antigen.
39. The method of treatment of claim 36 or 38, wherein the vessel is an artery. 40. The method of treatment of claim 36 or 38, wherein the vessel is a vein.
41. A medical device coated with at least one layer of a matrix comprising a fullerene ranging from about C60 to about CIOO.
42. The medical device of claim 41, wherein the first layer of the matrix is noncovalently attached to the medical device. 43. The medical device of claim 41, wherein the first layer of the matrix is covalently attached to the medical device.
44. The medical device of claim 43, wherein the matrix is C60O.
45. The medical device of claim 41, wherein the medical device is a stent.
46. The medical device of claim 41, wherein the medical device is a synthetic graft. 47. A medical device coated with a matrix consisting of a fullerene aπanged as a nanotube.
48. The medical device of claim 47, wherein the matrix is noncovalently attached to the medical device.
49. The medical device of claim 47, wherein the matrix is covalently attached to the medical device. 50. The medical device of claim 47, wherein the medical device is a stent.
51. The medical device of claim 47, wherein the medical device is a synthetic graft.
PCT/US2001/008244 2000-03-15 2001-03-15 Coating that promotes endothelial cell adherence WO2001068158A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CA002400319A CA2400319C (en) 2000-03-15 2001-03-15 Coating that promotes endothelial cell adherence
KR1020027012043A KR100860860B1 (en) 2000-03-15 2001-03-15 Coating that promotes endothelial cell adherence
JP2001566720A JP5859179B2 (en) 2000-03-15 2001-03-15 Coating that promotes endothelial cell adhesion
IL15150101A IL151501A0 (en) 2000-03-15 2001-03-15 Coating that promotes endothelial cell adherence
EP01918685A EP1263484B1 (en) 2000-03-15 2001-03-15 Coating which promotes endothelial cell adherence
DE60128451T DE60128451T2 (en) 2000-03-15 2001-03-15 COATING THAT STIMULATES A PLUG OF ENDOTHELIAL CELLS
AU2001245734A AU2001245734A1 (en) 2000-03-15 2001-03-15 Coating that promotes endothelial cell adherence

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US18967400P 2000-03-15 2000-03-15
US60/189,674 2000-03-15
US20178900P 2000-05-04 2000-05-04
US60/201,789 2000-05-04

Publications (1)

Publication Number Publication Date
WO2001068158A1 true WO2001068158A1 (en) 2001-09-20

Family

ID=26885401

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/008244 WO2001068158A1 (en) 2000-03-15 2001-03-15 Coating that promotes endothelial cell adherence

Country Status (12)

Country Link
US (6) US7037332B2 (en)
EP (1) EP1263484B1 (en)
JP (3) JP5859179B2 (en)
KR (1) KR100860860B1 (en)
CN (1) CN100506293C (en)
AT (1) ATE362382T1 (en)
AU (1) AU2001245734A1 (en)
CA (1) CA2400319C (en)
DE (1) DE60128451T2 (en)
ES (1) ES2283398T3 (en)
IL (1) IL151501A0 (en)
WO (1) WO2001068158A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003092763A1 (en) * 2002-05-03 2003-11-13 Duke University Carbon nanotubules for storage of nitric oxide
EP1471853A2 (en) * 2002-02-06 2004-11-03 Orbus Medical Technologies, Inc. Medical device with coating that promotes endothelial cell adherence and differentiation
JP2004344469A (en) * 2003-05-23 2004-12-09 Terumo Corp Medical device for embedding in living body
WO2005113033A3 (en) * 2004-05-20 2006-03-02 Boston Scient Scimed Inc Medical devices and methods of making the same
WO2006071297A1 (en) * 2004-12-22 2006-07-06 Boston Scientific Limited Vulnerable plaque stent
EP1740219A2 (en) * 2004-04-30 2007-01-10 OrbusNeich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods of using same
WO2006112932A3 (en) * 2005-02-17 2007-03-29 Advanced Cardiovascular System Coatings for implantable medical devices containing attractants for endothelial cells
WO2007087687A1 (en) * 2006-02-03 2007-08-09 University Of Wollongong Biocompatible composites
WO2007097887A2 (en) * 2006-02-15 2007-08-30 Advanced Cardiovascular Systems, Inc. Coatings for implantable medical devices containing attractants for endothelial cells
US7371425B2 (en) 2003-05-16 2008-05-13 Cinvention Ag Method for coating substrates with a carbon-based material
WO2008063157A2 (en) * 2006-10-25 2008-05-29 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services A nanoparticle-based anticoagulant
US7591831B2 (en) 2001-09-28 2009-09-22 Boston Scientific Scimed, Inc. Medical devices comprising nanocomposites
US8147859B2 (en) 2002-09-26 2012-04-03 Advanced Bio Prosthetic Surfaces, Ltd. Implantable material having patterned surface of raised elements and photochemically altered elements and method of making same
US8268340B2 (en) 2002-09-26 2012-09-18 Advanced Bio Prosthetic Surfaces, Ltd. Implantable materials having engineered surfaces and method of making same
US8361140B2 (en) 2009-12-29 2013-01-29 Boston Scientific Scimed, Inc. High strength low opening pressure stent design
ITVI20110310A1 (en) * 2011-11-29 2013-05-30 Ehsan Bahrami COMBINATORIAL COATING INCLUDING TWO CHEMICAL PROTEINS TO INCREASE THE ATTRACTION OF ENDOTELIAL CELLS AND ENDOTHIAL PROGENITRIC CELLS ON A METALLIC SURFACE
US8632583B2 (en) 2011-05-09 2014-01-21 Palmaz Scientific, Inc. Implantable medical device having enhanced endothelial migration features and methods of making the same
US8679517B2 (en) 2002-09-26 2014-03-25 Palmaz Scientific, Inc. Implantable materials having engineered surfaces made by vacuum deposition and method of making same
EP2716307A1 (en) * 2006-06-13 2014-04-09 Alchimedics Drug eluting stent with a biodegradable release layer attached with an electro-grafted primer coating
US8728563B2 (en) 2011-05-03 2014-05-20 Palmaz Scientific, Inc. Endoluminal implantable surfaces, stents, and grafts and method of making same
US9522217B2 (en) 2000-03-15 2016-12-20 Orbusneich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods for using same
WO2019211345A1 (en) * 2018-05-03 2019-11-07 Acandis Gmbh Medical device and coating with bonding agent
CN115052639A (en) * 2020-02-06 2022-09-13 信越化学工业株式会社 Angiogenesis device

Families Citing this family (203)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2178541C (en) 1995-06-07 2009-11-24 Neal E. Fearnot Implantable medical device
US6638237B1 (en) * 1999-08-04 2003-10-28 Percardia, Inc. Left ventricular conduits and methods for delivery
US7220276B1 (en) * 2000-03-06 2007-05-22 Surmodics, Inc. Endovascular graft coatings
US20030229393A1 (en) * 2001-03-15 2003-12-11 Kutryk Michael J. B. Medical device with coating that promotes cell adherence and differentiation
US8088060B2 (en) 2000-03-15 2012-01-03 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
AU2001245734A1 (en) * 2000-03-15 2001-09-24 Orbus Medical Technologies Inc. Coating that promotes endothelial cell adherence
US8460367B2 (en) * 2000-03-15 2013-06-11 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20070141107A1 (en) * 2000-03-15 2007-06-21 Orbusneich Medical, Inc. Progenitor Endothelial Cell Capturing with a Drug Eluting Implantable Medical Device
US9320829B2 (en) * 2000-03-15 2016-04-26 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20050271701A1 (en) * 2000-03-15 2005-12-08 Orbus Medical Technologies, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20070055367A1 (en) * 2000-03-15 2007-03-08 Orbus Medical Technologies, Inc. Medical device with coating that promotes endothelial cell adherence and differentiation
US6854467B2 (en) * 2000-05-04 2005-02-15 Percardia, Inc. Methods and devices for delivering a ventricular stent
US20020032478A1 (en) * 2000-08-07 2002-03-14 Percardia, Inc. Myocardial stents and related methods of providing direct blood flow from a heart chamber to a coronary vessel
US20060177416A1 (en) 2003-10-14 2006-08-10 Medivas, Llc Polymer particle delivery compositions and methods of use
US8192484B2 (en) 2000-12-12 2012-06-05 Cardiatis S.A. Stent for blood flow improvement
US6976990B2 (en) * 2001-01-25 2005-12-20 Percardia, Inc. Intravascular ventriculocoronary bypass via a septal passageway
DE10122806A1 (en) * 2001-05-10 2002-11-14 Holger Kiesewetter Method for the detection of blood cell antigens and antibodies directed against them
US7923055B2 (en) 2001-05-11 2011-04-12 Exogenesis Corporation Method of manufacturing a drug delivery system
US8889169B2 (en) * 2001-05-11 2014-11-18 Exogenesis Corporation Drug delivery system and method of manufacturing thereof
US7666462B2 (en) * 2001-05-11 2010-02-23 Exogenesis Corporation Method of controlling a drug release rate
WO2003002243A2 (en) 2001-06-27 2003-01-09 Remon Medical Technologies Ltd. Method and device for electrochemical formation of therapeutic species in vivo
US8252040B2 (en) 2001-07-20 2012-08-28 Microvention, Inc. Aneurysm treatment device and method of use
US7572288B2 (en) * 2001-07-20 2009-08-11 Microvention, Inc. Aneurysm treatment device and method of use
US8715312B2 (en) 2001-07-20 2014-05-06 Microvention, Inc. Aneurysm treatment device and method of use
US20030036698A1 (en) * 2001-08-16 2003-02-20 Robert Kohler Interventional diagnostic catheter and a method for using a catheter to access artificial cardiac shunts
WO2003037400A2 (en) * 2001-10-31 2003-05-08 Ventrigraft Inc Methods and device compositions for the recruitment of cells to blood contacting surfaces in vivo
US20030109824A1 (en) * 2001-11-07 2003-06-12 Microvena Corporation Distal protection device with local drug delivery to maintain patency
WO2003063575A2 (en) 2002-01-29 2003-08-07 Colb A Mark Endothelialization of vascular surfaces
US7008397B2 (en) * 2002-02-13 2006-03-07 Percardia, Inc. Cardiac implant and methods
US7758889B1 (en) 2002-07-24 2010-07-20 Luna Innovations Incorporated Fullerenes in targeted therapies
US20080193490A1 (en) * 2002-02-14 2008-08-14 Andreas Hirsch Use of Carbon Nanotube for Drug Delivery
AU2003217833A1 (en) * 2002-03-01 2003-09-16 Mark A. Colb Binding of red blood cell to exposed subendothelial surfaces to impede platelet deposition thereon and/or for use in targeted drug delivery thereto
US6905667B1 (en) * 2002-05-02 2005-06-14 Zyvex Corporation Polymer and method for using the polymer for noncovalently functionalizing nanotubes
EP1364663A1 (en) * 2002-05-21 2003-11-26 Commonwealth Scientific And Industrial Research Organisation Ocular devices with functionalized surface with adhesive properties
WO2004002367A1 (en) * 2002-06-27 2004-01-08 Microport Medical (Shanghai) Co., Ltd. Drug eluting stent
US7162308B2 (en) 2002-11-26 2007-01-09 Wilson Greatbatch Technologies, Inc. Nanotube coatings for implantable electrodes
US7060684B1 (en) 2002-12-16 2006-06-13 Quijano Rodolfo C Device for treating diabetes and methods thereof
US7311727B2 (en) * 2003-02-05 2007-12-25 Board Of Trustees Of The University Of Arkansas Encased stent
AU2004215898A1 (en) * 2003-02-26 2004-09-10 Medivas, Llc Bioactive stents and methods for use thereof
US7579077B2 (en) * 2003-05-05 2009-08-25 Nanosys, Inc. Nanofiber surfaces for use in enhanced surface area applications
US20050221072A1 (en) * 2003-04-17 2005-10-06 Nanosys, Inc. Medical device applications of nanostructured surfaces
US20060122596A1 (en) * 2003-04-17 2006-06-08 Nanosys, Inc. Structures, systems and methods for joining articles and materials and uses therefor
US7056409B2 (en) * 2003-04-17 2006-06-06 Nanosys, Inc. Structures, systems and methods for joining articles and materials and uses therefor
US7074294B2 (en) * 2003-04-17 2006-07-11 Nanosys, Inc. Structures, systems and methods for joining articles and materials and uses therefor
US20050038498A1 (en) * 2003-04-17 2005-02-17 Nanosys, Inc. Medical device applications of nanostructured surfaces
US7972616B2 (en) 2003-04-17 2011-07-05 Nanosys, Inc. Medical device applications of nanostructured surfaces
CA2522866A1 (en) * 2003-04-28 2005-01-20 Nanosys, Inc. Super-hydrophobic surfaces, methods of their construction and uses therefor
TWI427709B (en) * 2003-05-05 2014-02-21 Nanosys Inc Nanofiber surfaces for use in enhanced surface area applications
US7803574B2 (en) 2003-05-05 2010-09-28 Nanosys, Inc. Medical device applications of nanostructured surfaces
WO2005016414A2 (en) * 2003-08-13 2005-02-24 The Children's Hospital Of Philadelphia Use of receptor sequences for immobilizing gene vectors on surfaces
US20050124896A1 (en) * 2003-08-25 2005-06-09 Jacob Richter Method for protecting implantable sensors and protected implantable sensors
US20060188486A1 (en) * 2003-10-14 2006-08-24 Medivas, Llc Wound care polymer compositions and methods for use thereof
US20050096509A1 (en) * 2003-11-04 2005-05-05 Greg Olson Nanotube treatments for internal medical devices
US8333798B2 (en) 2003-11-07 2012-12-18 Merlin Md Pte Ltd. Implantable medical devices with enhanced visibility, mechanical properties and biocompatability
US20060085062A1 (en) * 2003-11-28 2006-04-20 Medlogics Device Corporation Implantable stent with endothelialization factor
US20090118817A1 (en) * 2005-06-16 2009-05-07 Mayo Foundation For Medical Education And Research Magnetic Medical Apparatus, Kits, and Methods
US8465453B2 (en) * 2003-12-03 2013-06-18 Mayo Foundation For Medical Education And Research Kits, apparatus and methods for magnetically coating medical devices with living cells
JP4618997B2 (en) * 2003-12-09 2011-01-26 テルモ株式会社 Stent and manufacturing method thereof
US20050153309A1 (en) 2003-12-22 2005-07-14 David Hoon Method and apparatus for in vivo surveillance of circulating biological components
US20110039690A1 (en) 2004-02-02 2011-02-17 Nanosys, Inc. Porous substrates, articles, systems and compositions comprising nanofibers and methods of their use and production
US8025960B2 (en) 2004-02-02 2011-09-27 Nanosys, Inc. Porous substrates, articles, systems and compositions comprising nanofibers and methods of their use and production
US20050175667A1 (en) * 2004-02-10 2005-08-11 Wenda Carlyle Use of endothelin antagonists to prevent restenosis
CA2555364C (en) * 2004-03-10 2012-10-16 Orbus Medical Technologies, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US8715340B2 (en) * 2004-03-31 2014-05-06 Merlin Md Pte Ltd. Endovascular device with membrane
SG133420A1 (en) * 2005-12-13 2007-07-30 Merlin Md Pte Ltd An endovascular device with membrane having permanently attached agents
US8915952B2 (en) 2004-03-31 2014-12-23 Merlin Md Pte Ltd. Method for treating aneurysms
US8500751B2 (en) 2004-03-31 2013-08-06 Merlin Md Pte Ltd Medical device
US8163269B2 (en) * 2004-04-05 2012-04-24 Carpenter Kenneth W Bioactive stents for type II diabetics and methods for use thereof
US20060013855A1 (en) * 2004-04-05 2006-01-19 Medivas, Llc Bioactive stents for type II diabetics and methods for use thereof
US20050234545A1 (en) * 2004-04-19 2005-10-20 Yea-Yang Su Amorphous oxide surface film for metallic implantable devices and method for production thereof
WO2005114194A1 (en) * 2004-05-21 2005-12-01 Mds Inc. Doing Business Through Its Mds Pharma Services Division Method of quantifying the cell-binding properties of a medical device
AU2005251814A1 (en) * 2004-06-07 2005-12-22 Innovational Holdings, Llc Local delivery of growth factors for stem cell transplantation
EP1771214A2 (en) * 2004-06-16 2007-04-11 Affinergy Inc. Biofunctional coatings
US20060093642A1 (en) * 2004-11-03 2006-05-04 Ranade Shrirang V Method of incorporating carbon nanotubes in a medical appliance, a carbon nanotube medical appliance, and a medical appliance coated using carbon nanotube technology
GB2420976B (en) * 2004-11-19 2006-12-20 Zvi Finkelstein Therapeutic implant
US8609123B2 (en) * 2004-11-29 2013-12-17 Advanced Cardiovascular Systems, Inc. Derivatized poly(ester amide) as a biobeneficial coating
US20080118561A1 (en) * 2004-12-08 2008-05-22 Helen Marie Nugent Materials and Methods for Minimally-Invasive Administration of a Cell-Containing Flowable Composition
US20070100321A1 (en) * 2004-12-22 2007-05-03 Leon Rudakov Medical device
US8158410B2 (en) * 2005-01-18 2012-04-17 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
JP2008532643A (en) * 2005-03-09 2008-08-21 ザ ユニバーシティー オブ テネシー リサーチ ファウンデーション Barrier stent and use thereof
WO2006116357A1 (en) * 2005-04-21 2006-11-02 Massachusetts Institute Of Technology Materials and methods for altering an immune response to exogenous and endogenous immunogens, including syngeneic and non-syngeneic cells, tissues or organs
US8414907B2 (en) * 2005-04-28 2013-04-09 Warsaw Orthopedic, Inc. Coatings on medical implants to guide soft tissue healing
US9119901B2 (en) * 2005-04-28 2015-09-01 Warsaw Orthopedic, Inc. Surface treatments for promoting selective tissue attachment to medical impants
ES2263382B1 (en) * 2005-05-16 2007-11-16 Fundacion Para La Investigacion Biomedica Del Hospital Gregorio Marañon ARTIFICIAL MATRIX OF ENDOTHELIZED FIBRINE GEL SUPERPRODUCTOR OF PROANGIOGEN FACTORS.
JP5632580B2 (en) * 2005-06-21 2014-11-26 シャイア リジェネラティブ メディシン, インコーポレイテッド Methods and compositions for improving vascular access
CA2623239C (en) * 2005-09-22 2016-07-12 Medivas, Llc Solid polymer delivery compositions and methods for use thereof
JP5192384B2 (en) 2005-09-22 2013-05-08 メディバス エルエルシー Bis- (α-amino) -diol-diester-containing poly (ester amide) and poly (ester urethane) compositions and methods of use
US8900620B2 (en) 2005-10-13 2014-12-02 DePuy Synthes Products, LLC Drug-impregnated encasement
US20070196423A1 (en) * 2005-11-21 2007-08-23 Med Institute, Inc. Implantable medical device coatings with biodegradable elastomer and releasable therapeutic agent
US20100204783A1 (en) * 2005-12-06 2010-08-12 Helen Marie Nugent Methods and compositions for enhancing vascular access
EP1962894A4 (en) * 2005-12-07 2012-11-14 Medivas Llc Method for assembling a polymer-biologic delivery composition
US8840660B2 (en) 2006-01-05 2014-09-23 Boston Scientific Scimed, Inc. Bioerodible endoprostheses and methods of making the same
US7807624B2 (en) * 2006-01-11 2010-10-05 Affinergy, Inc. Methods and compositions for promoting attachment of cells of endothelial cell lineage to medical devices
US7531505B2 (en) * 2006-01-11 2009-05-12 Affinergy, Inc. Compositions and methods for promoting attachment of cells of endothelial cell lineage to medical devices
US8089029B2 (en) 2006-02-01 2012-01-03 Boston Scientific Scimed, Inc. Bioabsorbable metal medical device and method of manufacture
US20070191766A1 (en) * 2006-02-10 2007-08-16 Boston Scientific Scimed, Inc. Balloon catheter having nanotubes
JP4750860B2 (en) * 2006-02-13 2011-08-17 マーリン エムディー ピーティーイー リミテッド Intravascular device having a membrane
US20070203564A1 (en) * 2006-02-28 2007-08-30 Boston Scientific Scimed, Inc. Biodegradable implants having accelerated biodegradation properties in vivo
US20070225795A1 (en) * 2006-03-24 2007-09-27 Juan Granada Composite vascular prosthesis
US8048150B2 (en) 2006-04-12 2011-11-01 Boston Scientific Scimed, Inc. Endoprosthesis having a fiber meshwork disposed thereon
EP2019645A4 (en) * 2006-05-02 2013-03-06 Medivas Llc Delivery of ophthalmologic agents to the exterior or interior of the eye
JP5196498B2 (en) * 2006-05-09 2013-05-15 メディバス エルエルシー Biodegradable water-soluble polymer
US7572625B2 (en) * 2006-05-18 2009-08-11 Boston Scientific Scimed, Inc. Medical devices coated with drug carrier macromolecules
US20080108138A1 (en) * 2006-06-13 2008-05-08 Vermette Patrick Bioactive compositions and their use in cell patterning
US8535372B1 (en) * 2006-06-16 2013-09-17 Abbott Cardiovascular Systems Inc. Bioabsorbable stent with prohealing layer
US8066753B2 (en) * 2006-07-06 2011-11-29 Robert Kipperman Specialized catheter and method for placement in a bifurcated vessel
US7824438B2 (en) * 2006-07-06 2010-11-02 Robert Kipperman Method for placement of a stent assembly in a bifurcated vessel
WO2008017028A2 (en) 2006-08-02 2008-02-07 Boston Scientific Scimed, Inc. Endoprosthesis with three-dimensional disintegration control
US20080085293A1 (en) * 2006-08-22 2008-04-10 Jenchen Yang Drug eluting stent and therapeutic methods using c-Jun N-terminal kinase inhibitor
EP2089072B1 (en) 2006-09-06 2014-06-04 Boston Scientific Limited Medical devices having a coating for promoting endothelial cell adhesion
CA2663220A1 (en) 2006-09-15 2008-03-20 Boston Scientific Limited Medical devices and methods of making the same
WO2008034048A2 (en) 2006-09-15 2008-03-20 Boston Scientific Limited Bioerodible endoprosthesis with biostable inorganic layers
ATE517590T1 (en) 2006-09-15 2011-08-15 Boston Scient Ltd BIOLOGICALLY ERODABLE ENDOPROTHESES
US8808726B2 (en) 2006-09-15 2014-08-19 Boston Scientific Scimed. Inc. Bioerodible endoprostheses and methods of making the same
JP2010503482A (en) 2006-09-18 2010-02-04 ボストン サイエンティフィック リミテッド Endoprosthesis
WO2008039521A2 (en) * 2006-09-26 2008-04-03 Nmt Medical, Inc. Method for modifying a medical implant surface for promoting tissue growth
JP5372764B2 (en) * 2006-11-07 2013-12-18 シャイア リジェネラティブ メディシン, インコーポレイテッド Materials and methods for the treatment and management of angiogenesis-related diseases
CA2674195A1 (en) 2006-12-28 2008-07-10 Boston Scientific Limited Bioerodible endoprostheses and methods of making same
CN114377204A (en) 2007-01-30 2022-04-22 匹兹堡大学 Bioerodible coverings and uses thereof
US20080311172A1 (en) * 2007-04-25 2008-12-18 Schapira Jay N Programmed-release, nanostructured biological construct
WO2008151130A1 (en) * 2007-06-01 2008-12-11 Ev3 Peripheral, Inc. Extension tubes for balloon catheters
US20100070020A1 (en) 2008-06-11 2010-03-18 Nanovasc, Inc. Implantable Medical Device
ES2381641T3 (en) * 2007-06-13 2012-05-30 Pervasis Therapeutics, Inc. Methods and devices for minimally invasive administration of fluid compositions containing cells
CA2704920C (en) 2007-06-25 2016-08-16 Microvention, Inc. Self-expanding prosthesis
EP2178944A1 (en) * 2007-07-24 2010-04-28 Medivas, LLC Biodegradable cationic polymer gene transfer compositions and methods of use
US20090043330A1 (en) * 2007-08-09 2009-02-12 Specialized Vascular Technologies, Inc. Embolic protection devices and methods
US20090043380A1 (en) * 2007-08-09 2009-02-12 Specialized Vascular Technologies, Inc. Coatings for promoting endothelization of medical devices
US8052745B2 (en) 2007-09-13 2011-11-08 Boston Scientific Scimed, Inc. Endoprosthesis
JP5356239B2 (en) 2007-09-28 2013-12-04 テルモ株式会社 In vivo indwelling
CN101172168B (en) * 2007-10-10 2010-06-02 大连理工大学 Metallic blood vessel bracket coating for osamine glycan load CD133 antibody and method for preparing the same
US20090112239A1 (en) * 2007-10-31 2009-04-30 Specialized Vascular Technologies, Inc. Sticky dilatation balloon and methods of using
US8319002B2 (en) 2007-12-06 2012-11-27 Nanosys, Inc. Nanostructure-enhanced platelet binding and hemostatic structures
CN101883545B (en) 2007-12-06 2013-08-07 纳诺西斯有限公司 Resorbable nanoenhanced hemostatic structures and bandage materials
US8128706B2 (en) * 2008-01-09 2012-03-06 Innovative Health Technologies, Llc Implant pellets and methods for performing bone augmentation and preservation
JP5435878B2 (en) * 2008-02-12 2014-03-05 富士フイルム株式会社 Labeled particles with fragmented antibody immobilized on labeling substance
WO2009114089A2 (en) * 2008-03-03 2009-09-17 Luna Innovations Incorporated Using fullerenes to enhance and stimulate hair growth
WO2009114090A2 (en) * 2008-03-03 2009-09-17 Luna Innovations Incorporated Method for inhibiting the build-up of arterial plaque by administering fullerenes
US7998192B2 (en) 2008-05-09 2011-08-16 Boston Scientific Scimed, Inc. Endoprostheses
US8236046B2 (en) 2008-06-10 2012-08-07 Boston Scientific Scimed, Inc. Bioerodible endoprosthesis
US20100022940A1 (en) * 2008-07-25 2010-01-28 Medtronic Vascular, Inc. Percutaneously Introduceable Shunt Devices and Methods
US7985252B2 (en) 2008-07-30 2011-07-26 Boston Scientific Scimed, Inc. Bioerodible endoprosthesis
WO2010017456A2 (en) * 2008-08-07 2010-02-11 Exogenesis Corporation Drug delivery system and method of munufacturing thereof
JP5878369B2 (en) * 2008-08-07 2016-03-08 エクソジェネシス コーポレーション Medical device for bone implant and method of manufacturing the device
WO2010019716A1 (en) * 2008-08-13 2010-02-18 Medivas, Llc Aabb-poly(depsipeptide) biodegradable polymers and methods of use
US8642063B2 (en) 2008-08-22 2014-02-04 Cook Medical Technologies Llc Implantable medical device coatings with biodegradable elastomer and releasable taxane agent
US8262692B2 (en) * 2008-09-05 2012-09-11 Merlin Md Pte Ltd Endovascular device
US8382824B2 (en) 2008-10-03 2013-02-26 Boston Scientific Scimed, Inc. Medical implant having NANO-crystal grains with barrier layers of metal nitrides or fluorides
WO2010054121A2 (en) * 2008-11-07 2010-05-14 Specialized Vascular Technologies, Inc. Extracellular matrix modulating coatings for medical devices
US8540889B1 (en) 2008-11-19 2013-09-24 Nanosys, Inc. Methods of generating liquidphobic surfaces
US8267992B2 (en) 2009-03-02 2012-09-18 Boston Scientific Scimed, Inc. Self-buffering medical implants
US8574612B2 (en) * 2009-03-04 2013-11-05 Boston Scientific Scimed, Inc. Medical devices having a coating of biologic macromolecules
US9205177B2 (en) * 2009-03-04 2015-12-08 Peytant Solutions, Inc. Stents modified with material comprising amnion tissue and corresponding processes
WO2010132707A1 (en) * 2009-05-14 2010-11-18 Orbusneich Medical, Inc. Self-expanding stent with polygon transition zone
US8366763B2 (en) 2009-07-02 2013-02-05 Tryton Medical, Inc. Ostium support for treating vascular bifurcations
EP2464392A4 (en) * 2009-08-12 2013-04-10 Univ Duke Cell coated implantable device
ES2669694T3 (en) 2009-10-19 2018-05-28 The Population Council, Inc. Neuroprotection and myelin repair using Nestorone®
US9446051B2 (en) * 2009-10-19 2016-09-20 The Population Council, Inc. Neuroprotection and myelin repair using nestorone®
JP2013509258A (en) 2009-10-28 2013-03-14 ユニバーシティ オブ ピッツバーグ オブ ザ コモンウェルス システム オブ ハイヤー エデュケイション Bioerodible wrap and uses therefor
WO2011084559A2 (en) 2009-12-16 2011-07-14 Neograft Technologies, Inc. Graft devices and methods of use
US9295541B2 (en) 2009-12-31 2016-03-29 Neograft Technologies, Inc. Graft devices and methods of fabrication
WO2011119573A1 (en) 2010-03-23 2011-09-29 Boston Scientific Scimed, Inc. Surface treated bioerodible metal endoprostheses
US8579964B2 (en) 2010-05-05 2013-11-12 Neovasc Inc. Transcatheter mitral valve prosthesis
WO2012012407A2 (en) 2010-07-19 2012-01-26 Neograft Technologies, Inc. Graft devices and methods of use
US9861814B2 (en) * 2010-12-23 2018-01-09 Medtronic, Inc. Medical electrical lead having biological surface and methods of making and using same
KR101770827B1 (en) * 2011-03-10 2017-08-23 서울대학교산학협력단 Stent for selectively capturing epc and process for producing the same
US9308087B2 (en) 2011-04-28 2016-04-12 Neovasc Tiara Inc. Sequentially deployed transcatheter mitral valve prosthesis
US9554897B2 (en) 2011-04-28 2017-01-31 Neovasc Tiara Inc. Methods and apparatus for engaging a valve prosthesis with tissue
US10058330B2 (en) 2011-05-11 2018-08-28 Microvention, Inc. Device for occluding a lumen
US9873765B2 (en) 2011-06-23 2018-01-23 Dsm Ip Assets, B.V. Biodegradable polyesteramide copolymers for drug delivery
CA2839841C (en) 2011-06-23 2019-11-05 Dsm Ip Assets B.V. New biodegradable polyesteramide copolymers for drug delivery
WO2013016349A2 (en) 2011-07-25 2013-01-31 Neograft Technologies, Inc. Vessel treatment methods and devices for use in a graft device
GB201114293D0 (en) 2011-08-19 2011-10-05 Univ Edinburgh Endothelial polymers
US9381112B1 (en) 2011-10-06 2016-07-05 William Eric Sponsell Bleb drainage device, ophthalmological product and methods
US8632489B1 (en) 2011-12-22 2014-01-21 A. Mateen Ahmed Implantable medical assembly and methods
TWI590843B (en) 2011-12-28 2017-07-11 信迪思有限公司 Films and methods of manufacture
JP6324371B2 (en) 2012-04-06 2018-05-16 マーリン エムディー プライベート リミテッド Devices and methods for treating aneurysms
EP2841024B1 (en) 2012-04-26 2017-05-03 Tryton Medical, Inc. Support for treating vascular bifurcations
US9345573B2 (en) 2012-05-30 2016-05-24 Neovasc Tiara Inc. Methods and apparatus for loading a prosthesis onto a delivery system
US9572665B2 (en) 2013-04-04 2017-02-21 Neovasc Tiara Inc. Methods and apparatus for delivering a prosthetic valve to a beating heart
BR112015032045B1 (en) 2013-06-21 2020-06-09 Depuy Synthes Products Inc flexible body, method for forming a multilayer film for use in combination with an implantable medical device, film storage system and orthopedic treatment system
CN104383609B (en) * 2014-11-07 2016-09-14 浙江大学 The application in screening endotheliocyte and smooth muscle cell of the material of a kind of controllable cell adhesion
US10888442B2 (en) * 2014-11-11 2021-01-12 University of Pittsburgh—of the Commonwealth System of Higher Education Super elastic, bioabsorbable endovascular devices
CA2969171C (en) 2014-12-18 2023-12-12 Dsm Ip Assets B.V. Drug delivery system for delivery of acid sensitive drugs
EP3389735B1 (en) 2015-12-19 2022-03-23 Cardiac Pacemakers, Inc. Biologically inert coating for implantable medical devices
CN108882981B (en) 2016-01-29 2021-08-10 内奥瓦斯克迪亚拉公司 Prosthetic valve for preventing outflow obstruction
SG11201810969RA (en) 2016-06-09 2019-01-30 Haimachek Inc Collector for detection and reversible capturing of cells from body fluids in vivo
WO2017218787A1 (en) 2016-06-16 2017-12-21 Cardiac Pacemakers, Inc. Hydrophilization and antifouling of enhanced metal surfaces
EP3496771B1 (en) 2016-08-09 2023-01-04 Cardiac Pacemakers, Inc. Functionalized peg for implantable medical devices
US20180206851A1 (en) * 2016-10-19 2018-07-26 Daniel E. Walzman Hydrogel intrasaccular occlusion device
CN109996581B (en) 2016-11-21 2021-10-15 内奥瓦斯克迪亚拉公司 Methods and systems for rapid retrieval of transcatheter heart valve delivery systems
US10603485B2 (en) 2016-11-28 2020-03-31 Boston Scientific Neuromodulation Corporation Features in increased surface area on neuromodulation leads
CN108619581A (en) * 2017-03-21 2018-10-09 微创心脉医疗科技(上海)有限公司 The application of medication coat, interventional medical device and epiphysin in medication coat
EP3609434A4 (en) * 2017-04-13 2020-11-25 OrbusNeich Medical Pte. Ltd. Medical devices coated with polydopamine and antibodies
US10433852B2 (en) 2017-05-08 2019-10-08 William Z. H'Doubler Aortic occlusion balloon apparatus, system and method of making
CN111263622A (en) 2017-08-25 2020-06-09 内奥瓦斯克迪亚拉公司 Sequentially deployed transcatheter mitral valve prosthesis
JP2021534906A (en) * 2018-08-31 2021-12-16 ウィスコンシン アラムニ リサーチ ファンデーション Generation of vascular grafts seeded with arterial endothelial cells
EP3876870B1 (en) 2018-11-08 2023-12-20 Neovasc Tiara Inc. Ventricular deployment of a transcatheter mitral valve prosthesis
EP3946163A4 (en) 2019-04-01 2022-12-21 Neovasc Tiara Inc. Controllably deployable prosthetic valve
EP3952792A4 (en) 2019-04-10 2023-01-04 Neovasc Tiara Inc. Prosthetic valve with natural blood flow
CN114025813A (en) 2019-05-20 2022-02-08 内奥瓦斯克迪亚拉公司 Introducer with hemostatic mechanism
WO2020257643A1 (en) 2019-06-20 2020-12-24 Neovasc Tiara Inc. Low profile prosthetic mitral valve
CN110917410B (en) * 2019-11-14 2020-11-24 浙江大学 Cardiovascular stent coating based on double-layer heterogeneous structure and preparation method thereof
CN113713172B (en) * 2021-09-08 2023-04-11 深圳清华大学研究院 In-situ endothelialization promoting coating and preparation method thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5310669A (en) * 1992-06-22 1994-05-10 The Trustees Of Dartmouth College Fullerene coated surfaces and uses thereof
WO1997032571A1 (en) * 1996-03-06 1997-09-12 Hyperion Catalysis International, Inc. Functionalized nanotubes
WO1998022541A2 (en) * 1996-11-08 1998-05-28 Ikonos Corporation Method for coating substrates
WO1999032184A1 (en) * 1997-12-19 1999-07-01 Cordis Corporation Catheter system having fullerenes and method
WO1999036276A1 (en) * 1998-01-16 1999-07-22 Etex Corporation Surface modification of medical implants

Family Cites Families (160)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3523807A (en) 1966-11-25 1970-08-11 Mihaly Gerendas Method of making a cross-linked fibrin prosthesis
JPS5313915B2 (en) 1973-01-25 1978-05-13
US4515160A (en) 1982-04-23 1985-05-07 Medtronic, Inc. Cardiac pacemaker synchronized programming
US4487715A (en) 1982-07-09 1984-12-11 The Regents Of The University Of California Method of conjugating oligopeptides
US4548736A (en) 1983-08-29 1985-10-22 Wisconsin Alumni Research Foundation Preparation of protein films
US4553974A (en) 1984-08-14 1985-11-19 Mayo Foundation Treatment of collagenous tissue with glutaraldehyde and aminodiphosphonate calcification inhibitor
US4889808A (en) * 1985-10-01 1989-12-26 American Home Products Method of enchancing t-PA and SCU-PA production
JPS62235568A (en) * 1985-12-05 1987-10-15 ドクタ− ミユ−ラ−−リ−ルハイム ケ−ジ− バイオロジツシエ ラボラトリ−ン Carrier biochemically activated by antibody covalent-bonded with surface
US4962091A (en) 1986-05-23 1990-10-09 Syntex (U.S.A.) Inc. Controlled release of macromolecular polypeptides
US4920016A (en) 1986-12-24 1990-04-24 Linear Technology, Inc. Liposomes with enhanced circulation time
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US4795459A (en) 1987-05-18 1989-01-03 Rhode Island Hospital Implantable prosthetic device with lectin linked endothelial cells
US4886062A (en) 1987-10-19 1989-12-12 Medtronic, Inc. Intravascular radially expandable stent and method of implant
DE68923423T2 (en) 1988-10-28 1996-01-25 Terumo Corp Antithromotic medical material, artificial internal organ and method for producing an antithrombotic medical material.
US5043165A (en) 1988-12-14 1991-08-27 Liposome Technology, Inc. Novel liposome composition for sustained release of steroidal drugs
US5011778A (en) * 1989-05-23 1991-04-30 Otsuka Pharmaceutical Co., Ltd. Monoclonal antibodies directed to IL-1 activated endothelial cells and medicaments employing the monoclonal antibodies
US5635386A (en) 1989-06-15 1997-06-03 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture
JPH0366384A (en) 1989-08-04 1991-03-22 Senjiyu Seiyaku Kk System for controlling release of physiologically active material
US5674848A (en) 1989-08-14 1997-10-07 The Regents Of The University Of California Bioreactor compositions with enhanced cell binding
US5635482A (en) 1989-08-14 1997-06-03 The Regents Of The University Of California Synthetic compounds and compositions with enhanced cell binding
US5059166A (en) 1989-12-11 1991-10-22 Medical Innovative Technologies R & D Limited Partnership Intra-arterial stent with the capability to inhibit intimal hyperplasia
US5674192A (en) * 1990-12-28 1997-10-07 Boston Scientific Corporation Drug delivery
US5199939B1 (en) 1990-02-23 1998-08-18 Michael D Dake Radioactive catheter
WO1991012779A1 (en) 1990-02-28 1991-09-05 Medtronic, Inc. Intralumenal drug eluting prosthesis
US7189410B1 (en) * 1990-11-27 2007-03-13 The American National Red Cross Supplemented and unsupplemented tissue sealants, methods of their production and use
US5492890A (en) * 1990-12-03 1996-02-20 The Scripps Research Institute Polypeptides for promoting cell attachment
DE4202282A1 (en) * 1991-01-29 1992-08-20 Fuji Photo Film Co Ltd LIGHT SENSITIVE TRANSMISSION MATERIAL AND PICTURE PRODUCTION METHOD
US5198263A (en) 1991-03-15 1993-03-30 The United States Of America As Represented By The United States Department Of Energy High rate chemical vapor deposition of carbon films using fluorinated gases
US5199942A (en) 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
US5302168A (en) 1991-09-05 1994-04-12 Hess Robert L Method and apparatus for restenosis treatment
US5811447A (en) * 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6515009B1 (en) * 1991-09-27 2003-02-04 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5464450A (en) 1991-10-04 1995-11-07 Scimed Lifesystems Inc. Biodegradable drug delivery vascular stent
GB9203037D0 (en) * 1992-02-11 1992-03-25 Salutar Inc Contrast agents
US5302166A (en) * 1992-02-21 1994-04-12 Optima Industries, Inc. Tool changer
US6306421B1 (en) * 1992-09-25 2001-10-23 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5292813A (en) 1992-10-02 1994-03-08 Exxon Research & Engineering Co. Fullerene-grafted polymers and processes of making
US5981568A (en) * 1993-01-28 1999-11-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5338571A (en) 1993-02-10 1994-08-16 Northwestern University Method of forming self-assembled, mono- and multi-layer fullerene film and coated substrates produced thereby
US5464650A (en) 1993-04-26 1995-11-07 Medtronic, Inc. Intravascular stent and method
US5641466A (en) 1993-06-03 1997-06-24 Nec Corporation Method of purifying carbon nanotubes
US5558903A (en) 1993-06-10 1996-09-24 The Ohio State University Method for coating fullerene materials for tribology
US6120764A (en) 1993-06-24 2000-09-19 Advec, Inc. Adenoviruses for control of gene expression
AU7515294A (en) 1993-07-28 1995-02-28 Fred Hutchinson Cancer Research Center Creating novel hematopoietic cell lines by expressing altered retinoic acid receptors
US5543328A (en) * 1993-08-13 1996-08-06 Genetic Therapy, Inc. Adenoviruses having modified fiber proteins
CA2170357A1 (en) 1993-08-25 1995-03-02 David Digiusto Method for producing a highly enriched population of hematopoietic stem cells
US6323184B1 (en) * 1993-10-15 2001-11-27 Thomas Jefferson University Arteriovenous and venous graft treatments: methods and compositions
EP0754064B1 (en) 1994-04-08 2003-05-28 Atrix Laboratories, Inc. An adjunctive polymer system for use with medical device
US5536641A (en) 1994-05-17 1996-07-16 Memorial Sloane Kittering Cancer Center Monoclonal antibody specific for vascular endothelial cell antigen endoglyx-1 and uses thereof for detection of, and isolation of, vascular endothelial cells
GB9413029D0 (en) 1994-06-29 1994-08-17 Common Services Agency Stem cell immobilisation
US6249703B1 (en) 1994-07-08 2001-06-19 Medtronic, Inc. Handheld patient programmer for implantable human tissue stimulator
AU700584C (en) 1994-08-12 2002-03-28 Meadox Medicals, Inc. Vascular graft impregnated with a heparin-containing collagen sealant
US6342390B1 (en) * 1994-11-23 2002-01-29 The United States Of America As Represented By The Secretary Of Health And Human Services Lipid vesicles containing adeno-associated virus rep protein for transgene integration and gene therapy
US6281015B1 (en) 1994-12-16 2001-08-28 Children's Medical Center Corp. Localized delivery of factors enhancing survival of transplanted cells
US6231600B1 (en) 1995-02-22 2001-05-15 Scimed Life Systems, Inc. Stents with hybrid coating for medical devices
WO1996029998A1 (en) 1995-03-28 1996-10-03 Fidia Advanced Biopolymers S.R.L. Nanospheres comprising a biocompatible polysaccharide
US6120536A (en) * 1995-04-19 2000-09-19 Schneider (Usa) Inc. Medical devices with long term non-thrombogenic coatings
US6099562A (en) * 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US5807398A (en) * 1995-04-28 1998-09-15 Shaknovich; Alexander Shuttle stent delivery catheter
US5780436A (en) 1995-05-01 1998-07-14 The Regents Of The University Of California Peptide compositions with growth factor-like activity
US5661127A (en) 1995-05-01 1997-08-26 The Regents Of The University Of California Peptide compositions with growth factor-like activity
US5603722A (en) 1995-06-06 1997-02-18 Quanam Medical Corporation Intravascular stent
US5824299A (en) * 1995-06-22 1998-10-20 President & Fellows Of Harvard College Modulation of endothelial cell proliferation with IP-10
US5830879A (en) * 1995-10-02 1998-11-03 St. Elizabeth's Medical Center Of Boston, Inc. Treatment of vascular injury using vascular endothelial growth factor
US6090618A (en) * 1996-10-07 2000-07-18 Arch Development Corporation DNA constructs and viral vectors comprising a smooth muscle promoter
US5669924A (en) * 1995-10-26 1997-09-23 Shaknovich; Alexander Y-shuttle stent assembly for bifurcating vessels and method of using the same
DK2111876T3 (en) * 1995-12-18 2011-12-12 Angiodevice Internat Gmbh Crosslinked polymer preparations and methods for their use
US5865723A (en) 1995-12-29 1999-02-02 Ramus Medical Technologies Method and apparatus for forming vascular prostheses
WO1997029802A2 (en) 1996-02-20 1997-08-21 Advanced Bionics Corporation Improved implantable microstimulator and systems employing the same
US20040072768A1 (en) * 1996-03-07 2004-04-15 Khachigian Levon Michael Inhibition of proliferation of cells
AUPN855496A0 (en) * 1996-03-07 1996-04-04 Unisearch Limited Prevention of proliferation of vascular cells
JPH09249751A (en) * 1996-03-14 1997-09-22 Collagn Corp Use of hydrophobic crosslinking agent for preparing crosslinked biological material composition
US6790606B1 (en) * 1996-03-15 2004-09-14 Munin Corporation Extracellular matrix signaling molecules
WO1997033995A2 (en) * 1996-03-15 1997-09-18 Munin Corporation Extracellular matrix signalling molecules
GB9606188D0 (en) 1996-03-23 1996-05-29 Danbiosyst Uk Pollysaccharide microspheres for the pulmonary delivery of drugs
GB9606452D0 (en) 1996-03-27 1996-06-05 Sandoz Ltd Organic compounds
US5843633A (en) 1996-04-26 1998-12-01 Amcell Corporation Characterization of a human hematopoietic progenitor cell antigen
US6455678B1 (en) 1996-04-26 2002-09-24 Amcell Corporation Human hematopoietic stem and progenitor cell antigen
US6143037A (en) * 1996-06-12 2000-11-07 The Regents Of The University Of Michigan Compositions and methods for coating medical devices
US6013780A (en) * 1996-09-06 2000-01-11 Technion Research & Development Co. Ltd. VEGF145 expression vectors
WO1998017796A2 (en) 1996-10-25 1998-04-30 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Methods and compositions for inhibiting inflammation and angiogenesis comprising a mammalian cd97 alpha subunit
US6500421B1 (en) 1996-11-04 2002-12-31 St. Jude Children's Research Hospital In vivo selection of primitive hematopoietic cells
US5980887A (en) 1996-11-08 1999-11-09 St. Elizabeth's Medical Center Of Boston Methods for enhancing angiogenesis with endothelial progenitor cells
US5749890A (en) * 1996-12-03 1998-05-12 Shaknovich; Alexander Method and system for stent placement in ostial lesions
US5753088A (en) 1997-02-18 1998-05-19 General Motors Corporation Method for making carbon nanotubes
US6258939B1 (en) 1997-03-10 2001-07-10 The Regents Of The University Of California PSCA antibodies and hybridomas producing them
US6238872B1 (en) * 1997-04-01 2001-05-29 S.E.T.-Smart Endolumenal Technologies Ltd. Targeted therapy to a biomedical device
US20020032315A1 (en) * 1997-08-06 2002-03-14 Manuel Baca Anti-vegf antibodies
US6273913B1 (en) 1997-04-18 2001-08-14 Cordis Corporation Modified stent useful for delivery of drugs along stent strut
US6077987A (en) 1997-09-04 2000-06-20 North Shore-Long Island Jewish Research Institute Genetic engineering of cells to enhance healing and tissue regeneration
US5880090A (en) * 1997-09-19 1999-03-09 The Hope Heart Institute Treatment of vascular graft implants with G-CSF
US6440734B1 (en) 1998-09-25 2002-08-27 Cytomatrix, Llc Methods and devices for the long-term culture of hematopoietic progenitor cells
US8257725B2 (en) * 1997-09-26 2012-09-04 Abbott Laboratories Delivery of highly lipophilic agents via medical devices
US5945457A (en) * 1997-10-01 1999-08-31 A.V. Topchiev Institute Of Petrochemical Synthesis, Russian Academy Of Science Process for preparing biologically compatible polymers and their use in medical devices
US5980565A (en) 1997-10-20 1999-11-09 Iowa-India Investments Company Limited Sandwich stent
US20020056148A1 (en) 1997-11-14 2002-05-09 Readhead Carol W. Transfection, storage and transfer of male germ cells for generation of selectable transgenic stem cells
WO1999025863A1 (en) 1997-11-14 1999-05-27 Cedars-Sinai Medical Center Transfection and transfer of male germ cells for generation of transgenic species
US6197586B1 (en) 1997-12-12 2001-03-06 The Regents Of The University Of California Chondrocyte-like cells useful for tissue engineering and methods
US20020051762A1 (en) 1998-01-23 2002-05-02 Shahin Rafii Purified populations of endothelial progenitor cells
US6541116B2 (en) 1998-01-30 2003-04-01 Advanced Cardiovascular Systems, Inc. Superoxide dismutase or superoxide dismutase mimic coating for an intracorporeal medical device
US20030125615A1 (en) 1998-02-05 2003-07-03 Yitzhack Schwartz Homing of autologous cells to a target zone in tissue using active therapeutics or substances
US6140127A (en) 1998-02-18 2000-10-31 Cordis Corporation Method of coating an intravascular stent with an endothelial cell adhesive five amino acid peptide
US6676937B1 (en) 1998-03-09 2004-01-13 Caritas St. Elizabeth's Medical Center Of Boston Inc. Compositions and methods for modulating vascularization
US6482406B1 (en) 1999-03-26 2002-11-19 Duncan J. Stewart Cell-based gene therapy for the pulmonary system
CA2329010A1 (en) 1998-04-17 1999-10-28 Angiogenix Incorporated Therapeutic angiogenic factors and methods for their use
US6206914B1 (en) 1998-04-30 2001-03-27 Medtronic, Inc. Implantable system with drug-eluting cells for on-demand local drug delivery
US6153252A (en) 1998-06-30 2000-11-28 Ethicon, Inc. Process for coating stents
AU5087799A (en) * 1998-07-06 2000-01-24 Beth Israel Deaconess Medical Center Methods of inhibiting proliferative diseases by inhibiting tgf-beta mediated angiogenesis
CA2279476C (en) 1998-07-31 2011-09-27 Stemcell Technologies Inc. Novel antibody composition for isolating human cells from human-murine chimeric hematopoietic cell suspensions
US6387663B1 (en) * 1998-07-31 2002-05-14 University Of Southern California Targeting pharmaceutical agents to injured tissues
JP2000086699A (en) 1998-09-07 2000-03-28 Seikagaku Kogyo Co Ltd New monoclonal antibody for recognizing cell surface antigen cd14
DE69911629D1 (en) * 1998-10-08 2003-10-30 Childrens Hospital Boston COMPOSITIONS AND THEIR USE FOR INHIBITING ANGIOGENESIS
US6375680B1 (en) * 1998-12-01 2002-04-23 St. Jude Medical, Inc. Substrates for forming synthetic tissues
EP1149111B1 (en) * 1999-01-06 2009-08-12 University Of Southern California Method and composition for angiogenesis inhibition
DE19903385A1 (en) 1999-01-29 2000-08-03 Max Delbrueck Centrum Agent for the prevention of in-stent restenosis and post-operative inflammation
US6544541B1 (en) * 1999-06-02 2003-04-08 Cardiovascular Solutions, Inc. Devices and compounds for treating arterial restenosis
JP2003501092A (en) * 1999-06-07 2003-01-14 エドワーズ ライフサイエンシーズ コーポレイション Targeted angiogenesis
US6258121B1 (en) 1999-07-02 2001-07-10 Scimed Life Systems, Inc. Stent coating
US6159531A (en) 1999-08-30 2000-12-12 Cardiovasc, Inc. Coating having biological activity and medical implant having surface carrying the same and method
US6371980B1 (en) 1999-08-30 2002-04-16 Cardiovasc, Inc. Composite expandable device with impervious polymeric covering and bioactive coating thereon, delivery apparatus and method
US6503273B1 (en) * 1999-11-22 2003-01-07 Cyograft Tissue Engineering, Inc. Tissue engineered blood vessels and methods and apparatus for their manufacture
US20010053362A1 (en) * 2000-03-09 2001-12-20 Lee Walters Applications of immune system tolerance to treatment of various diseases
US20070141107A1 (en) * 2000-03-15 2007-06-21 Orbusneich Medical, Inc. Progenitor Endothelial Cell Capturing with a Drug Eluting Implantable Medical Device
US20030229393A1 (en) 2001-03-15 2003-12-11 Kutryk Michael J. B. Medical device with coating that promotes cell adherence and differentiation
US9320829B2 (en) * 2000-03-15 2016-04-26 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US8088060B2 (en) * 2000-03-15 2012-01-03 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US8460367B2 (en) * 2000-03-15 2013-06-11 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20160287708A9 (en) * 2000-03-15 2016-10-06 Orbusneich Medical, Inc. Progenitor Endothelial Cell Capturing with a Drug Eluting Implantable Medical Device
US20050271701A1 (en) 2000-03-15 2005-12-08 Orbus Medical Technologies, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
AU2001245734A1 (en) * 2000-03-15 2001-09-24 Orbus Medical Technologies Inc. Coating that promotes endothelial cell adherence
US9522217B2 (en) 2000-03-15 2016-12-20 Orbusneich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods for using same
HUP0300810A2 (en) * 2000-07-20 2003-08-28 M.G.V.S. Ltd. Artifical vascular grafts, and methods of producing and using same
US6607720B1 (en) 2000-09-05 2003-08-19 Yong-Fu Xiao Genetically altered mammalian embryonic stem cells, their living progeny, and their therapeutic application for improving cardiac function after myocardial infarction
US6548025B1 (en) 2000-11-08 2003-04-15 Illinois Institute Of Technology Apparatus for generating odor upon electronic signal demand
US6471980B2 (en) 2000-12-22 2002-10-29 Avantec Vascular Corporation Intravascular delivery of mycophenolic acid
US6613083B2 (en) * 2001-05-02 2003-09-02 Eckhard Alt Stent device and method
US7666393B2 (en) 2001-05-11 2010-02-23 Children's Medical Center Corporation Methods for assessing antiangiogenic agents
WO2002096195A1 (en) 2001-05-31 2002-12-05 Enteromed, Inc. Treatment or replacement therapy using transgenic stem cells delivered to the gut
DE10148828B4 (en) * 2001-10-04 2005-05-19 Avontec Gmbh Modulation of the expression of STAT-1-dependent genes
WO2003037400A2 (en) * 2001-10-31 2003-05-08 Ventrigraft Inc Methods and device compositions for the recruitment of cells to blood contacting surfaces in vivo
WO2003063575A2 (en) * 2002-01-29 2003-08-07 Colb A Mark Endothelialization of vascular surfaces
AUPS078002A0 (en) * 2002-02-27 2002-03-21 Unisearch Limited Dnazyme therapeutics
US20030181973A1 (en) 2002-03-20 2003-09-25 Harvinder Sahota Reduced restenosis drug containing stents
JP4136434B2 (en) 2002-04-17 2008-08-20 進 清野 Induction of insulin-producing cells
CN1655738A (en) 2002-05-20 2005-08-17 奥勃斯医学技术股份有限公司 Drug eluting implantable medical device
EP1410812A1 (en) 2002-10-14 2004-04-21 EC (European Community) Functionalisation of stents with lipase
PL376752A1 (en) * 2002-11-07 2006-01-09 Abbott Laboratories Prosthesis having varied concentration of beneficial agent
AU2003296663A1 (en) * 2002-12-18 2004-07-09 Novartis Ag Endothelial cell specifically binding peptides
US7186882B2 (en) * 2003-03-18 2007-03-06 Nasrin Mesaeli Transgenic mouse over-expressing calreticulin (CRT) in vascular smooth muscle cells
US20050107870A1 (en) * 2003-04-08 2005-05-19 Xingwu Wang Medical device with multiple coating layers
US20050149174A1 (en) 2003-12-18 2005-07-07 Medtronic Vascular, Inc. Medical devices to treat or inhibit restenosis
US20050187607A1 (en) * 2004-02-20 2005-08-25 Akhtar Adil J. Drug delivery device
US8431145B2 (en) * 2004-03-19 2013-04-30 Abbott Laboratories Multiple drug delivery from a balloon and a prosthesis
EP1735042B1 (en) * 2004-03-19 2011-11-23 Abbott Laboratories Multiple drug delivery from a balloon and a prosthesis
WO2005107817A2 (en) 2004-04-30 2005-11-17 Orbus Medical Technologies, Inc. Medical device with coating for capturing genetically-altered cells and methods of using same
US7959942B2 (en) * 2006-10-20 2011-06-14 Orbusneich Medical, Inc. Bioabsorbable medical device with coating
EP2073754A4 (en) * 2006-10-20 2012-09-26 Orbusneich Medical Inc Bioabsorbable polymeric composition and medical device background
US8026382B2 (en) * 2007-05-18 2011-09-27 Heidi Kay Lipid raft, caveolin protein, and caveolar function modulation compounds and associated synthetic and therapeutic methods
EP2281002A2 (en) * 2008-04-25 2011-02-09 MorphoSys AG Anti-alk1 antibodies and uses thereof
US9217042B2 (en) * 2012-10-23 2015-12-22 Abbott Cardiovascular Systems Inc. Method of reducing MACE in diabetic patients subsequent to stent placement

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5310669A (en) * 1992-06-22 1994-05-10 The Trustees Of Dartmouth College Fullerene coated surfaces and uses thereof
WO1997032571A1 (en) * 1996-03-06 1997-09-12 Hyperion Catalysis International, Inc. Functionalized nanotubes
WO1998022541A2 (en) * 1996-11-08 1998-05-28 Ikonos Corporation Method for coating substrates
WO1999032184A1 (en) * 1997-12-19 1999-07-01 Cordis Corporation Catheter system having fullerenes and method
WO1999036276A1 (en) * 1998-01-16 1999-07-22 Etex Corporation Surface modification of medical implants

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE BIOSIS [online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1991, DEKKER A ET AL: "IMPROVED ADHESION AND PROLIFERATION OF HUMAN ENDOTHELIAL CELLS ON POLYETHYLENE PRECOATED WITH MONOCLONAL ANTIBODIES DIRECTED AGAINST CELL MEMBRANE ANTIGENS AND EXTRACELLULAR MATRIX PROTEINS", XP002172219, Database accession no. PREV199293037123 *
THROMBOSIS AND HAEMOSTASIS, vol. 66, no. 6, 1991, pages 715 - 724, ISSN: 0340-6245 *

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9364565B2 (en) 2000-03-15 2016-06-14 Orbusneich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods of using same
US9522217B2 (en) 2000-03-15 2016-12-20 Orbusneich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods for using same
US8133250B2 (en) 2001-09-28 2012-03-13 Boston Scientific Scimed, Inc. Medical devices comprising nanocomposites
US7591831B2 (en) 2001-09-28 2009-09-22 Boston Scientific Scimed, Inc. Medical devices comprising nanocomposites
US9463103B2 (en) 2001-09-28 2016-10-11 Boston Scientific Scimed, Inc. Medical devices comprising nanocomposites
EP1471853A2 (en) * 2002-02-06 2004-11-03 Orbus Medical Technologies, Inc. Medical device with coating that promotes endothelial cell adherence and differentiation
JP2005523050A (en) * 2002-02-06 2005-08-04 オーバス メディカル テクノロジーズ インク. Medical device having a coating that promotes endothelial cell adhesion and differentiation
EP1471853A4 (en) * 2002-02-06 2010-03-24 Orbusneich Medical Inc Medical device with coating that promotes endothelial cell adherence and differentiation
CN100455275C (en) * 2002-02-06 2009-01-28 祥丰医疗有限公司 Medical device with coating that promotes endothelial cell adherence and differentiation
WO2003092763A1 (en) * 2002-05-03 2003-11-13 Duke University Carbon nanotubules for storage of nitric oxide
US10314949B2 (en) 2002-09-26 2019-06-11 Vactronix Scientific, Llc Implantable biomaterials having functional surfaces
US10034967B2 (en) 2002-09-26 2018-07-31 Vactronix Scientific, Llc Implantable biomaterials having engineered functional surfaces
US9272077B2 (en) 2002-09-26 2016-03-01 Palmaz Scientific, Inc. Implantable materials having engineered surfaces and method of making same
US8932347B2 (en) 2002-09-26 2015-01-13 Advanced Bio Prosthetic Surfaces, Ltd. Implantable materials having engineered surfaces and method of making same
US10039866B2 (en) 2002-09-26 2018-08-07 Vactronix Scientific, Llc Implantable materials having engineered surfaces and method of making same
US8709066B2 (en) 2002-09-26 2014-04-29 Advanced Bio Prosthetic Surfaces, Ltd. Implantable materials having engineered surfaces comprising a pattern of features and method of making same
US8679517B2 (en) 2002-09-26 2014-03-25 Palmaz Scientific, Inc. Implantable materials having engineered surfaces made by vacuum deposition and method of making same
US10682443B2 (en) 2002-09-26 2020-06-16 Vactronix Scientific, Llc Implantable biomaterials having functional surfaces
US10729824B2 (en) 2002-09-26 2020-08-04 Vactronix Scientific, Llc. Implantable materials having engineered surfaces and method of making same
US8147859B2 (en) 2002-09-26 2012-04-03 Advanced Bio Prosthetic Surfaces, Ltd. Implantable material having patterned surface of raised elements and photochemically altered elements and method of making same
US8268340B2 (en) 2002-09-26 2012-09-18 Advanced Bio Prosthetic Surfaces, Ltd. Implantable materials having engineered surfaces and method of making same
US7371425B2 (en) 2003-05-16 2008-05-13 Cinvention Ag Method for coating substrates with a carbon-based material
JP2004344469A (en) * 2003-05-23 2004-12-09 Terumo Corp Medical device for embedding in living body
EP1740219A2 (en) * 2004-04-30 2007-01-10 OrbusNeich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods of using same
EP1740219A4 (en) * 2004-04-30 2009-01-28 Orbusneich Medical Inc Medical device with coating for capturing genetically-altered cells and methods of using same
EP2946666A1 (en) * 2004-04-30 2015-11-25 OrbusNeich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods of using same
WO2005113033A3 (en) * 2004-05-20 2006-03-02 Boston Scient Scimed Inc Medical devices and methods of making the same
WO2006071297A1 (en) * 2004-12-22 2006-07-06 Boston Scientific Limited Vulnerable plaque stent
WO2006112932A3 (en) * 2005-02-17 2007-03-29 Advanced Cardiovascular System Coatings for implantable medical devices containing attractants for endothelial cells
WO2007087687A1 (en) * 2006-02-03 2007-08-09 University Of Wollongong Biocompatible composites
WO2007097887A3 (en) * 2006-02-15 2007-10-18 Advanced Cardiovascular System Coatings for implantable medical devices containing attractants for endothelial cells
WO2007097887A2 (en) * 2006-02-15 2007-08-30 Advanced Cardiovascular Systems, Inc. Coatings for implantable medical devices containing attractants for endothelial cells
EP2716307A1 (en) * 2006-06-13 2014-04-09 Alchimedics Drug eluting stent with a biodegradable release layer attached with an electro-grafted primer coating
WO2008063157A2 (en) * 2006-10-25 2008-05-29 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services A nanoparticle-based anticoagulant
WO2008063157A3 (en) * 2006-10-25 2009-03-19 Us Gov Health & Human Serv A nanoparticle-based anticoagulant
US8361140B2 (en) 2009-12-29 2013-01-29 Boston Scientific Scimed, Inc. High strength low opening pressure stent design
US8728563B2 (en) 2011-05-03 2014-05-20 Palmaz Scientific, Inc. Endoluminal implantable surfaces, stents, and grafts and method of making same
US9439789B2 (en) 2011-05-09 2016-09-13 Palmaz Scientific, Inc. Implantable medical device having enhanced endothelial migration features and methods of making the same
US10206772B2 (en) 2011-05-09 2019-02-19 Vactronix Scientific, Llc Implantable medical device having enhanced endothelial migration features and methods of making the same
US8632583B2 (en) 2011-05-09 2014-01-21 Palmaz Scientific, Inc. Implantable medical device having enhanced endothelial migration features and methods of making the same
US10786343B2 (en) 2011-05-09 2020-09-29 Vactronix Scientific, Llc Implantable medical device having enhanced endothelial migration features and methods of making the same
ITVI20110310A1 (en) * 2011-11-29 2013-05-30 Ehsan Bahrami COMBINATORIAL COATING INCLUDING TWO CHEMICAL PROTEINS TO INCREASE THE ATTRACTION OF ENDOTELIAL CELLS AND ENDOTHIAL PROGENITRIC CELLS ON A METALLIC SURFACE
WO2019211345A1 (en) * 2018-05-03 2019-11-07 Acandis Gmbh Medical device and coating with bonding agent
EP4088756A1 (en) * 2018-05-03 2022-11-16 Acandis GmbH Medical device for use in human vessels having a fibrin coating including fibronectin
CN115052639A (en) * 2020-02-06 2022-09-13 信越化学工业株式会社 Angiogenesis device

Also Published As

Publication number Publication date
US7803183B2 (en) 2010-09-28
DE60128451D1 (en) 2007-06-28
US20150352263A1 (en) 2015-12-10
JP2003526477A (en) 2003-09-09
US20150352261A1 (en) 2015-12-10
EP1263484A1 (en) 2002-12-11
US7037332B2 (en) 2006-05-02
JP5859179B2 (en) 2016-02-10
ES2283398T3 (en) 2007-11-01
CN100506293C (en) 2009-07-01
JP2015128598A (en) 2015-07-16
ATE362382T1 (en) 2007-06-15
AU2001245734A1 (en) 2001-09-24
JP5675744B2 (en) 2015-02-25
CN1418115A (en) 2003-05-14
US20020049495A1 (en) 2002-04-25
CA2400319A1 (en) 2001-09-20
US9072723B2 (en) 2015-07-07
EP1263484B1 (en) 2007-05-16
US20130035755A1 (en) 2013-02-07
KR100860860B1 (en) 2008-09-29
DE60128451T2 (en) 2007-08-30
US9555166B2 (en) 2017-01-31
IL151501A0 (en) 2003-04-10
CA2400319C (en) 2008-09-16
JP2013046771A (en) 2013-03-07
US20050043787A1 (en) 2005-02-24
KR20030001378A (en) 2003-01-06
US20070156232A1 (en) 2007-07-05

Similar Documents

Publication Publication Date Title
US9555166B2 (en) Medical device with coating that promotes endothelial cell adherence
CA2472031C (en) Medical device with coating that promotes endothelial cell adherence and differentiation
EP2946666B1 (en) Medical device with coating for capturing genetically-altered cells and methods of using same
US9522217B2 (en) Medical device with coating for capturing genetically-altered cells and methods for using same
US20130172988A1 (en) Medical device with coating that promotes endothelial cell adherence and differentiation
US20060135476A1 (en) Medical device with coating that promotes endothelial cell adherence and differentiation
US20070191932A1 (en) Medical device with coating for capturing genetically-altered cells and methods for using same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2400319

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 151501

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 1020027012043

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 018066801

Country of ref document: CN

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 566720

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 2001918685

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001918685

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020027012043

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 2001918685

Country of ref document: EP