CA2673856A1 - Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain - Google Patents

Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain Download PDF

Info

Publication number
CA2673856A1
CA2673856A1 CA002673856A CA2673856A CA2673856A1 CA 2673856 A1 CA2673856 A1 CA 2673856A1 CA 002673856 A CA002673856 A CA 002673856A CA 2673856 A CA2673856 A CA 2673856A CA 2673856 A1 CA2673856 A1 CA 2673856A1
Authority
CA
Canada
Prior art keywords
dosage form
drug
sufentanil
dosage forms
sublingual
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
CA002673856A
Other languages
French (fr)
Other versions
CA2673856C (en
Inventor
Pamela Palmer
Thomas Schreck
Stelios Tzannis
Larry Hamel
Andrew I. Poutiatine
Charles Rampersaud
Bruce Edwards
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vertical Pharmaceuticals LLC
Original Assignee
Individual
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
Priority claimed from US11/650,174 external-priority patent/US8202535B2/en
Application filed by Individual filed Critical Individual
Publication of CA2673856A1 publication Critical patent/CA2673856A1/en
Application granted granted Critical
Publication of CA2673856C publication Critical patent/CA2673856C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/006Oral mucosa, e.g. mucoadhesive forms, sublingual droplets; Buccal patches or films; Buccal sprays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4535Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom, e.g. pizotifen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7007Drug-containing films, membranes or sheets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients

Abstract

Compositions, methods and systems for administration of small volume sufe ntanil-containing drug dosage forms to the oral mucosa of a subject are disc losed.

Description

SMALL VOLUME ORAL TRANSMUCOSAL DOSAGE FORMS CONTAINING
SUFENTANIL FOR TREATMENT OF PAIN

CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a continuation-in-part of U.S. Patent Application Serial No.
11/985,162, filed November 14, 2007; and U.S. Patent Application Serial No.
11/650,174, filed January 5, 2007, both of which are hereby incorporated by reference in their entirety.

Field of the Invention [0001] The invention relates to small volume sufentanil-containing drug dosage forms, together with methods and systems for oral transmucosal administration to a subject.

Background Of The Invention.
[0002] Oral dosage forms account for approximately eighty percent of all the drug dosage forms on the market. They are non-invasive, easily administered and have high patient compliance. Orally administered therapeutic agents, however, must be transported to the stomach and small intestine for absorption across the gastrointestinal (GI) mucosal membranes into the blood. The efficiency of absorption of a drug following oral administration can be low because of metabolism within the GI tract and first-pass metabolism within the liver, resulting in relatively lengthy onset times or erratic absorption characteristics that are not well-suited to control acute disorders. The majority of oral dosage forms on the market are designed for GI delivery. Relatively few oral dosage forms are designed for delivery through the oral mucosa.
[0003] Oral transmucosal delivery offers a number of advantages in that it can provide a shorter onset time to maximal plasma concentration (Cmax) than oral delivery, in particular for lipophilic drugs. This is because the drug rapidly passes directly and efficiently through the epithelium of the highly vascularized mucosal tissue to the plasma, thus rapidly reaching the circulation while avoiding slower, often inefficient and variable GI uptake. It is therefore advantageous for a drug to be delivered through the mucous membranes of the oral cavity, (e.g., via the sublingual route), when rapid onset, consistent Tmax and Cmax are advantageous.
[0004] In the process of oral transmucosal drug delivery, the drug is absorbed through the epithelial membranes of the oral cavity. However, frequently the key risk associated with oral transmucosal delivery is the enhanced potential for swallowing the medication owing to the continuous generation, backward flow and swallowing of the saliva. This becomes a particular risk when the dosage forms employed are large enough to produce a significant saliva response, which, in turn, leads to swallowing of drug and/or loss of adherence of the dosage form to the oral mucosa.
[0005] Various solid dosage forms, such as sublingual tablets, troches, lozenges, lozenges-on-a-stick, chewing gums, and buccal patches have been used to deliver drugs via the oral mucosal tissue. Solid dosage forms such as lozenges and tablets have been used for oral transmucosal delivery of drugs such as nitroglycerin sublingual tablets.
[0006] Reproducible and effective drug delivery technology represents an area of active research, in particular, as it applies to controlled substances such as opioids like sufentanil.
[0007] The relevant art does not describe a solid drug dosage form for delivery of sufentanil to the oral mucosa, e.g., the sublingual space.
[0008] Controlled access oral transmucosal drug dispensing systems offer numerous advantages over conventional means of drug administration such as oral and intravenous routes, the most important of which is enhanced safety, with additional advantages being rapid and consistent onset of action, more consistent and predictable plasma concentrations and higher and more consistent bioavailability than currently available dosage forms.
[0009] This is particularly relevant to the treatment of pain, more specifically, acute (i.e. post-operative), intermittent and breakthrough pain.
[0010] Therefore, a need exists for drug dosage forms, methods and systems for administration of an opioid, such as sufentanil (e.g., by patient-controlled administration), for treatment of pain, wherein the drug dosage form is administered with a device which provides for safe and controlled delivery of the drug via the oral mucosa, while minimizing the potential for drug abuse and/or diversion.
[0011] The present invention addresses these needs.

Brief Summary Of The Invention.
[0012] A single sublingual administration of the disclosed sufentanil dosage forms to a subject results in one or more of the following: a bioavailability of greater than 50%; an AUCinf with a coefficient of variation of less than 40%; a Tmax with a coefficient of variation of less than 40%; a linear relationship between Cmax and the amount of sufentanil in the dosage form; and a linear relationship between AUCinf and the amount of sufentanil in the dosage form.
[0013] Repeated sublingual administrations of the disclosed sufentanil dosage forms to a subject results in one or more of the following: a bioavailability which is greater than the bioavailability following a single sublingual administration to the subject; a difference between the Tmax following repeated sublingual administrations and the time of the previous sublingual administration which is shorter than the Tmax following a single sublingual administration to the subject; and a Tmax with a coefficient of variation of less than 40%.
[0014] The disclosed sufentanil dosage forms find utility in methods for treating pain.
[0015] Use of a dispensing device for placement of a sufentanil dosage form in the sublingual space in methods for treating pain is disclosed, wherein placement/

administration may be patient controlled.
[0016] The disclosed handheld dispensing devices comprise one or more of the following features: a housing having a dispensing end with a means to prevent or retard saliva ingress; a lock-out feature; a user identification feature; and a disposable cartridge configured to hold one or more drug dosage forms.
[0017] The lock-out feature may provide for repeated sublingual administration of sufentanil at a minimum interval of 20 minutes.
[0018] The cartridge may provide a smart cartridge recognition system comprising a physical keyed feature on the cartridge, an optically detected feature or pattern, a bar code on the cartridge, a magnetic tag on the cartridge, an RFID tag on the cartridge, an electronic microchip on the cartridge, or a combination thereof.
Brief Description Of The Drawings.
[0019] Figure 1 is a graphic depiction of sufentanil plasma concentration mean +/-standard deviation (SD) versus time, following sublingual administration of 2.5, 5, 10 and 20 mcg (5mcg every 10 minutes x 4 doses) sufentanil dosage forms (slow-eroding) in healthy human volunteers.
[0020] Figure 2 is a graphic depiction of the linearity of Cmax (mean +/- SD) versus sufentanil dose (mcg), following sublingual administration of 2.5, 5, 10 or 4 x 5mcg sufentanil dosage forms (slow-eroding) in healthy human volunteers.
[0021] Figure 3 is a graphic depiction of the linearity of AUC;nf (mean +/-SD) versus sufentanil dose (mcg), following sublingual administration of 2.5, 5, 10 or 4 x 5mcg sufentanil dosage forms (slow-eroding) in healthy human volunteers.
[0022] Figure 4 is a graphic depiction of sufentanil plasma concentration (mean +/- SD) versus time, following repeated sublingual administration of four 5mcg sufentanil dosage forms (slow-eroding) at 10 minute intervals in healthy human volunteers as compared to IV Infusion of 5 mcg sufentanil over 10 minutes.
[0023] Figures 5A and 5B provide a graphic depiction of observed and predicted sufentanil plasma concentration (mean +/- SD) versus time, following repeated sublingual administration of 4 x 5mcg sufentanil dosage forms (slow-eroding) at 10 minute intervals in healthy human volunteers over a period of 12 hours (Fig.
5A) or a period of 2.5 hours (Fig. 5B).
[0024] Figure 6 is a graphic depiction of sufentanil plasma concentrations (mean +/- SD) versus time, following sublingual administration of 10, 40 (10mcg every 20 minutes x 4 doses) and 80mcg sufentanil dosage forms (faster-eroding) in healthy human volunteers.
[0025] Figures 7A and 7B are a graphic depiction of observed and predicted sufentanil plasma concentration (mean +/- SD) versus time, following repeated sublingual administration of 4 x 10mcg sufentanil dosage forms (faster-eroding) at minute intervals in healthy human volunteers over a period of 12 hours (Fig.
7A) or a period of 2.5 hours (Fig. 7B).
[0026] Figure 8 is a graphic depiction of the linearity of Cmax (mean +/- SD) versus 20 sufentanil dose (mcg), following sublingual administration of 10, 4 x 10 or 80mcg sufentanil dosage forms (faster-eroding) in healthy human volunteers.
[0027] Figure 9 is a graphic depiction of the linearity of AUC;nf (mean +/-SD) versus sufentanil dose (mcg), following sublingual administration of 10, 4 x 10 or 80mcg sufentanil dosage forms (faster-eroding) in healthy human volunteers.
[0028] Figures 10A and 10B provide a graphic depiction of steady-state sufentanil plasma concentration versus time predicted by superposition following repeated sublingual administration of 10mcg doses of sufentanil at 20 minute intervals (Fig.
10A) or 15mcg doses of sufentanil at 20 minute intervals (Fig. 10B).
[0029] Figures 11A - E provide a schematic depiction of an exemplary dispensing device wherein the device is designed to deliver drug dosage forms to oral mucosa of a patient under treatment. Figs. 11A - E illustrate the progression of intact drug dispensing device 11 (Fig. 11A); the reusable head 13 and disposable body 15 of a drug dispensing device (Fig. 11B); a reusable head 13, disposable body 15 and cartridge 17, a dispense button 23, and a proboscis 31 of a drug dispensing device (Fig. 11C); various aspects of a drug dispensing device 11 including a reusable head 13, disposable body 15 and cartridge 17, a proboscis 31, and a latch 19 to unlock the device, a hub lock 21, a distal seal 33, 35, and a power train coupling 25 (Fig.
11D); and a reassembled intact drug dispensing device 11 (Fig. 11E).
[0030] Figure 12 is a schematic depiction of an exemplary dispensing device showing features designed to block or retard saliva and moisture ingress. The preferred embodiment includes a dispensing tip having a shroud 29, having one or more of: a wiping seal/valve 33, 35, an absorbent pad 39, a pushrod 51, a drying chamber/moisture communication channel 43, desiccant in the channel 45, a cartridge 17 containing dosage forms 67 and desiccant in the cartridge 47.
[0031] Figures 13A and 13B are schematic depictions of an exemplary geometry for a dispensing tip.
[0032] Figures 14A - D are a schematic depiction of an exemplary proboscis 31 of a dispensing device 11 wherein the proboscis 31 has an S-shape 53 and comprises a shroud 29 and a valve. The shroud shields the valve from moisture and saliva ingress from the tongue and other mucosa and provides an area for the dosage form to exit the device without "sticking" to the wetted distal valve or shroud area. The shroud also comprises a cut-out/ relief 55 in order to mitigate the dragging of dosage forms when the device is removed from the oral space. The valve functions with the shroud to control saliva and moisture ingress, as well as aid in delivery of the dosage form.
[0033] Figures 15A - D provide a series of flow diagrams for use of an exemplary device showing the stages of push rod/tablet interaction during device use, wherein Fig. 15A shows the LOAD feature; Fig. 15B shows the CALIBRATE feature; Fig.

shows the DISPENSE feature; and Fig. 15D shows the DISASSEMBLE feature.
[0034] Figure 16 is a schematic depiction of an exemplary device showing the stages of push rod/tablet interaction during device use. In Fig 16, the push rod 51, dosage forms 67, shipping tablet 69, spring 73 and position sensor 71 are shown.

During use, the push rod 51 moves between positions 57, 59, 61, 63, 65 and 67, also shown in Fig. 16.
[0035] Figure 17 is a schematic architecture connection diagram illustrating the various components that may be included in a drug dispensing device or system including a device with a separate drug dispensing device head 13, drug dispensing device body 15, drug cartridge 17, a portable docking FOB 113, a patient RFID
tag 115, and a base station 117.
[0036] Figure 18A is a block diagram illustrating one aspect of communication in the drug dispensing system, including an RFID tag, a drug dispensing device, a base station/dock and a healthcare provider personal computer.
[0037] Figure 18B is a block diagram illustrating another aspect of communication in a drug dispensing system, including an RFID tag, a drug dispensing device, a portable docking FOB, a base station and a healthcare provider personal computer.
[0038] Figures 19A and B are schematic depictions of an exemplary single dose applicator.
[0039] Figures 20A - C provide an illustration of one type of single dose applicator and use thereof in delivering a dosage form to a subject.
[0040] Figures 21A - F provide an illustration of six additional single dose applicators.
[0041] Figure 22 provides an illustration of a multiple dose applicator where a plurality of single dose applicators are stored prior to use.
[0042] Figures 23A - C provide an illustration of additional single dose applicator and multiple dose applicator embodiments.
[0043] Figures 24A - B provide an illustration of two stages of use of one embodiment of a single dose applicator.
[0044] Figs. 25A - D are schematic depictions of additional examples of single dose applicators (SDAs).
[0045] Figs. 26A - D provide a schematic depiction of a multiple dose applicator or container which provides for storage of a plurality of SDAs prior to use, and the use of the SDAs for sublingual administration of a drug dosage form.

Detailed Description Of The Invention.
I. Introduction
[0046] Provided herein are compositions, methods, systems, kits and drug dispensing devices for oral transmucosal, e.g., sublingual, administration of sufentanil-containing small volume dosage forms. Oral transmucosal administration of the dosage forms minimizes the saliva response and therefore minimizes delivery of the drug to the GI tract, such that the majority of drug is delivered across the oral mucosa. The small volume dosage forms have bioadhesive properties which facilitate adherence to the oral mucosa, thus minimizing the risk of ingestion and inefficient delivery due to swallowing.
[0047] The claimed small volume sufentanil-containing dosage forms, some embodiments of which are referred to as "Sublingual Sufentanil NanoTabsTM" , offer a number of advantages in terms of both safety and efficacy as compared to currently available pain treatments.
[0048] The following disclosure provides a description of the dosage forms, drug dispensing devices, methods, systems and kits which constitute the invention.
The invention is not limited to the specific dosage forms, devices, methodology, systems, kits or medical conditions described herein, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention.
[0049] It must be noted that as used herein and in the appended claims, the singular forms "a", "and", and "the" include plural references unless the context clearly dictates otherwise. Thus, for example, reference to "a drug formulation"
includes a plurality of such formulations and reference to "a drug delivery device"
includes systems comprising drug dosage forms and drug delivery devices for containment, storage and delivery of such dosage forms.
[0050] Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. Although any methods, devices and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, drug delivery devices and materials are now described.
[0051] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such a disclosure by virtue of prior invention.

II. Definitions
[0052] The term "active agent" or "active" may be used interchangeably herein with the term "drug" and is meant to refer to any therapeutically active agent.
[0053] The term "adhere" is used herein with reference to a drug dosage form or formulation that is in contact with a surface such as a mucosal surface and is retained on the surface without the application of an external force. The term "adhere" is not meant to imply any particular degree of sticking or bonding, nor is it meant to imply any degree of permanency.
[0054] The term "analgesic drug" as used herein includes sufentanil or a sufentanil congener, such as alfentanil, fentanyl, lofentanil, carfentanil, remifentanil, trefentanil, or mirfentanil, as well as formulations comprising one or more therapeutic compounds. Use of the phrase "sufentanil or a congener" is not meant to be limiting to use of, or formulations comprising, only one of these selected opioid compounds. Furthermore, reference to sufentanil alone or to a selected sufentanil congener alone, e.g., reference to "alfentanil", is understood to be only exemplary of the drugs suitable for delivery according to the methods of the invention, and is not meant to be limiting in any way.
[0055] The term "AUC" as used herein means "area under the curve", and is also referred to as"AUCinfin a plot of concentration of drug in plasma versus time.
AUC
is typically given for the time interval zero to infinity, however, clearly plasma drug concentrations cannot be measured 'to infinity' for a patient so a mathematical equation is used to estimate the AUC from a limited number of concentration measurements.
AUCinf= AUCt + Ciasdk where C,asf was the last plasma concentration.
[0056] In a practical sense, AUCinf represents the total amount of drug absorbed by the body, irrespective of the rate of absorption. This is useful when trying to determine whether two formulations of the same dose release the same dose of drug to the body. The AUCinf of a transmucosal dosage form compared to that of the same dosage administered intravenously serves as the basis for a measurement of bioavailability.
[0057] The term "bioadhesion" as used herein refers to adhesion to a biological surface including mucosal membranes.
[0058] The term "bioavailability" or "F" as used herein means "percent bioavailability" and represents the fraction of drug absorbed from a test article as compared to the same drug when administered intravenously. It is calculated from the AUCinf of the test article following delivery via the intended route versus the AUCinf for the same drug after intravenous administration. The absolute bioavailability of sublingual administration was determined by the following formula:

AUCsublingual Dose F(%) = mf x iV
7 A UCinf Dosesublingual
[0059] The term"breakthrough pain" as used herein, is a transitory flare of pain of moderate to severe intensity occurring on a background of otherwise controlled pain. "Breakthrough pain" can be intense for short periods of time, as short as 1 or 2 minutes or as long as 30 minutes or more.
[0060] The term "cartridge" is used herein with reference to a disposable cartridge configured to hold one or more drug dosage forms, typically, up to drug dosage forms. The cartridge may comprise a smart cartridge recognition system with a physical keyed feature on the cartridge, a bar code on the cartridge, a magnetic tag on the cartridge, an RFID tag on the cartridge, an electronic microchip on the cartridge, or a combination thereof. The cartridge may further comprise one or more shipping tablets wherein at least one shipping tablet is dispensed prior to dispensing of a dosage form.
[0061] The term "Cmax " as used herein means the maximum observed plasma concentration following administration of a drug.
[0062] The term "congener" as used herein refers to one of many variants or configurations of a common chemical structure.
[0063] The term "disintegration" is used interchangeably herein with "erosion"
and means the physical process by which a dosage form breaks down and pertains to the physical integrity of the dosage form alone. This can occur in a number of different ways including breaking into smaller pieces and ultimately, fine and large particulates or, alternatively, eroding from the outside in, until the dosage form has disappeared.
[0064] The term "dissolution" as used herein means the process by which the active ingredient is dissolved from the tablet in the presence of a solvent, in vitro, or physiological fluids in vivo, e.g., saliva, irrespective of the mechanism of release, diffusion, erosion or combined erosion and diffusion.
[0065] The term "dispensing device", "drug dispensing device", "dispenser", "drug dispenser", "drug dosage dispenser"," device" and "drug delivery device" are used interchangeably herein and refer to a device that dispenses a drug dosage form. The dispensing device provides for controlled and safe delivery of a pharmaceutically active substance (e.g., an opioid such as sufentanil) formulated in the dosage form.
The device may be adapted for storage and/or delivery of a dosage form such as a lozenge, pill, tablet, capsule, membrane, strip, liquid, patch, film, gel, spray or other form.
[0066] The term "dispensing end" as used herein with reference to a device means the portion of the device comprising the proboscis and shroud which serves to deliver a drug dosage form to the oral mucosa of a subject.
[0067] The term "drug", "medication", "pharmacologically active agent", "therapeutic agent" and the like are used interchangeably herein and generally refer to any substance that alters the physiology of an animal and can be effectively administered by the oral transmucosal route.
[0068] The term "erosion time" means the time required for a solid dosage form to break down until the dosage form has disappeared.
[0069] The term "FOB" refers to a small, portable handheld, powered electronic docking device that can be used in conjunction with the drug dispensing device to upload data, download data, control access to the drug dispensing device, control access to the drug dosage forms, or enhance or otherwise alter the user interface of the drug dispensing device. A FOB may communicate and dock with a drug dispensing device either in a wired or wireless fashion. A FOB may be adapted to attach to a cord so as to allow the FOB to hang from the neck of a healthcare professional such as a physician or caregiver, particularly in the hospital setting. A
drug dispensing device may communicate with the physician or care giver via the FOB.
[0070] The terms "formulation" and "drug formulation" as used herein refer to a physical composition containing at least one pharmaceutically active substance, which may be provided in any of a number of dosage forms for delivery to a subject.
The dosage form may be provided to the patient as a lozenge, pill, capsule, membrane, strip, liquid, patch, film, gum, gel, spray or other form.
[0071] The term "hydrogel-forming preparation", means a solid formulation largely devoid of water which upon contact with an aqueous solution, e.g., a bodily fluid, and in particular that of the oral mucosa, absorbs water in such a way that it forms a hydrated gel in situ. The formation of the gel follows unique disintegration (or erosion) kinetics while allowing for release of the therapeutic agent over time.
Additionally, the term "hydrogel-forming preparation' describes a solid formulation largely devoid of water which upon contact with bodily fluids, and in particular those in the oral cavity, transforms into a film that releases the drug. Such films increase the surface area available for drug release and absorption thus enabling faster absorption of the drug.
[0072] The term "lock-out feature" is used herein with reference to a feature of the device which provides for a "lock-out time".
[0073] The term "lock-out time" is used herein with reference to the period of time during which the device does not allow drug accessibility, i.e., a dosage form cannot be dispensed during the "lock-out time". "Lock-out time" may be programmable, a fixed time interval, a predetermined interval, a predetermined variable interval, an interval determined by an algorithm or a variable interval communicated to the device from a remote computer or docking station.
[0074] The term "LogP" as used herein means logarithm of the ratio of equilibrium concentrations of un-ionized compound between octanol and water. P
also called the'bctanol-water partition coefficient" and serves as a means to quantify the hydrophiobicity or lipophilicity of, a chemical characteristic of a given drug.
[0075] The term "mucoadhesion" is used herein in to refer to the adhesion to mucosal membranes which are covered by mucus, such as those in the oral cavity and may be used interchangeably herein with the term "bioadhesion" which refers to adhesion to any biological surface.
[0076] The term "mucosal membrane" refers generally to any of the mucus-coated biological membranes in the body. Absorption through the mucosal membranes of the oral cavity is of particular interest. Thus, oral mucosal absorption, i.e., buccal, sublingual, gingival and palatal absorption are specifically contemplated.
[0077] The term "mucosal-depot" is used herein in its broadest sense to refer to a reservoir or deposit of a pharmaceutically active substance within or just beneath the mucosal membrane.
[0078] The term "non-ordered particulate mixture" or "non-ordered mixture" is used herein with reference to a formulation where the mixture is not ordered with respect to the pharmaceutically active agent and the bioadhesive material or bioadhesion promoting agent, or other formulation components. In addition, it is used herein with reference to any formulation prepared by a process that involves dry mixing wherein drug particles are not uniformly distributed over the surface of larger carrier particles. Such `non-ordered' mixing may involve dry mixing of particles in a non-ordered fashion, where there is no requirement with respect to the order of addition/mixing of specific excipients with the drug, bioadhesive material or bioadhesion promoting agent and/or disintegrants. Further in the non-ordered mixing process, there is no limitation on the size of the drug particles. The drug particles may be larger than 25pm. In addition, a "non-ordered mixture"
includes any mixing processes in which the primary carrier particles do not incorporate a disintegrant within. Finally the "non-ordered mixture" may be prepared by any `wet mixing' processes, i.e. processes in which a solvent or non-solvent is added during the mixing process or any mixing process in which the drug is added in a solution or suspension form.
[0079] The term "operatively connected" as used herein means the components are provided in a device so as to function as intended to achieve an aim. For example, a memory device operatively connected to a CPU which is further operatively connected to a release mechanism may be meant to indicate that, upon actuation, the CPU communicates with the memory device to check the status or history of drug delivery, and then further communicates with the release mechanism (e.g., via a solenoid and a switch) to release and dispense a drug.
[0080] The term "opioid naive patient" is used herein with reference to a patient who has not received repeated administration of an opioid substance over a period of weeks to months.
[0081] The term "opioid tolerant patient" as used herein means a physiological state characterized by a decrease in the effects of an opioid substance (e.g., analgesia, nausea or sedation) with chronic administration. An opioid substance is a drug, hormone, or other chemical substance that has analgesic, sedative and/or narcotic effects similar to those containing opium or its derivatives. If analgesic tolerance develops, the dose of opioid substance is increased to result in the same level of analgesia. This tolerance may not extend to side effects and side effects may not be well tolerated as the dose is increased.
[0082] The terms "oral transmucosal dosage form" and "drug dosage form" may be used interchangeably herein and refer to a dosage form which comprises a pharmaceutically active substance, e.g., a drug such as sufentanil. The oral dosage form is used to deliver the pharmaceutically active substance to the circulation by way of the oral mucosa and is typically a "sublingual dosage form", but in some cases other oral transmucosal routes may be employed. The dosage form provides for delivery of the pharmaceutically active substance across the oral mucosa and by controlling the formulation the timing for release of the pharmaceutically active substance can be achieved. The dosage form comprises pharmaceutically acceptable excipients and may be referred to as a NanoTabT"', as detailed in U.S.
Application Serial No. 11/650,174. The dosage form comprises a formulation that is neither effervescent nor does it comprise an essentially water-free, ordered mixture of microparticles of drug adhered to the surface of carrier particles, where the carrier particles are substantially larger than the microparticles of drug.
[0083] The terms "oral transmucosal drug delivery" and "oral transmucosal administration" as used herein refer to drug delivery that occurs substantially via the oral transmucosal route and not via swallowing followed by GI absorption. This includes delivery via buccal, sublingual and gum transmucosal areas.
[0084] The term "proboscis" is used interchangeably with the terms "dispensing tip" a "delivery tip", and refers to a dispensing and/or positioning tip of a drug dosage form dispenser that delivers a dosage form to the oral mucosa (e.g., the sublingual space).
[0085] The term "radio frequency identification device" or "RFID" is used with reference to an automatic identification method, which relies on storing and remotely retrieving data using devices called RFID tags, wherein the RFID tag is applied to, or incorporated into a product, or person for the purpose of identification using radiowaves. Some tags can be read from several meters away and beyond the line of sight of the reader.
[0086] The term "replaceable cartridge" or "disposable cartridge" is used with reference to a cartridge for housing drug dosage forms which is typically configured to hold up to 200 drug dosage forms, wherein the cartridge is designed to be used and discarded.
[0087] The term "shipping tablet" is used herein with reference to an "initialization", or "shipping" tablet which is the same size and shape as a drug-containing dosage form but does not contain a pharmaceutically active substance.
The "shipping tablet" may comprise a placebo dosage form that does not contain a pharmaceutically active substance or may be made of plastic or other material.
It is the first thing dispensed from a new cartridge after insertion into a dispensing device. The device has a means for differentiating between the shipping tablet and a dosage form containing a pharmaceutically active substance.
[0088] The term "shroud" is used to describe a partial or complete covering of the dispensing end of the device which protects the delivery port from contact with saliva or other moisture in the oral cavity and forms a barrier between the device, the oral mucosa and tongue, has a relief for dosage form delivery, and an interior that is hydrophobic or hydrophilic which serves to minimize or eliminate saliva ingress or moisture ingress. The "shroud" creates a barrier from the oral mucosa contacting the valve area and dosage form, aiding in dosage form dispensing and discouraging dosage form adherence to the shroud. The shroud may have a rounded interior surface or other geometry to stop the dosage form adhering to the shroud. The shroud limits the ability of the tongue or oral mucosa to contact the dosage form dispensing area, thereby controlling saliva contact and ingress.
[0089] The term "subject" includes any subject, generally a mammal (e.g., human, canine, feline, equine, bovine, ungulate etc.), adult or child, in which treatment for a disorder is desired. The terms "subject" and "patient" may be used interchangeably herein.
[0090] The term "systems that include a drug dosage form and a dispensing device" as used herein refers to a drug dispensing system for delivery and/or monitoring of drug administration. The system may be used to monitor and deliver a pharmaceutically active substance, e.g., an opioid such as sufentanil, wherein the amount of drug delivered, corresponding efficacy and safety are enhanced over currently available systems. The system may have one or more features that provide for improved safety and ease of use over currently available systems including a security feature that prevents unauthorized access to the stored drugs, a dosing lock-out feature, a means for identifying an individual user for controlled drug access, a dose counting feature, a memory means for retaining information about dose delivery, and an interface for bidirectional exchange of information with a user, a drug cartridge, or another device such as a computer.
[0091] The term "small volume drug dosage form" or "small volume dosage form"
is used herein with reference to a small volume dosage form that has a volume of less than 100p1 and a mass of less than 100mg. More specifically, the dosage form has a mass of less than 100mg, 90mg, 80mg, 70mg, 60mg, 50mg, 40mg, 30mg, 29mg,28mg, 27mg, 26mg, 25mg, 24mg, 23mg, 22mg, 21mg, 20mg, 19mg,18mg, 17mg, 16mg, 15mg, 14mg, 13mg, 12mg, 11mg, 10mg, 9mg, 8mg, 7mg, 6mg or 5mg or a volume of less than 100p1, 90pl, 80pl, 70pl, 60 pl, 50pl, 40pl, 30pl, 29 pl, 28 pl, 27pl, 26pl, 25pl, 24pl, 23pl, 22pl, 21pl, 20pl, 19pl, 18pl, 17pl, 16pl, 15pl, 14pl, 13pl, 12pl, llpl, 10p1, 9pl, 8pl, 7pl, 6pl or 5pl. The "dosage form" may or may not have bioadhesive characteristics and may form a hydrogel upon contact with an aqueous solution.
[0092] The "dosage form" may be used to deliver any drug that can be administered by the oral transmucosal route in an amount amenable to administration via the small size of the dosage form, i.e. 0.25pg to 99.9mg, lpg to 50mg or lpg to 10mg.
[0093] The term "small volume sufentanil-containing drug dosage form" is used herein with reference to a small volume dosage form that contains a dose of sufentanil selected from about 2 micrograms (mcg) to about 200 mcg of sufentanil, e.g., 5 mcg, 10 mcg, 15 mcg, 20mcg, 30mcg, 40mcg, 50mcg, 60mcg, 70mcg, 80mcg or 100mcg of sufentanil.
[0094] The term "solid dosage form" or "solid drug dosage form" is used herein with reference to a small volume dosage form that is a solid, e.g., a lozenge, a pill, a tablet, a membrane or a strip.
[0095] The term "sublingual", means literally "under the tongue" and refers to administering a drug dosage form via the mouth in such a way that the pharmaceutically active substance is rapidly absorbed via the blood vessels under the tongue rather than via the digestive tract. Absorption occurs via the highly vascularized sublingual mucosa and allows the pharmaceutically active substance more direct access to the blood circulation, providing for direct systemic administration independent of GI influences.
[0096] The term "terminal half-life" or "ti/2 [h]" as defined herein is calculated as ln(2)/ AZ (defined as the first order terminal rate constant estimated by linear regression of the time versus log concentration curve) and also determined after the final dosing in repeated dose studies.
[0097] The term "Tmax " as used herein means the time point of maximum observed plasma concentration.
[0098] The term "Tonset " as used herein means the observed "time of onset"
and represents the time required for the plasma drug concentration to reach 50% of the maximum observed plasma concentration, Cmax=
[0099] The term "therapeutically effective amount" means an amount of a therapeutic agent, or a rate of delivery of a therapeutic agent (e.g., amount over time), effective to facilitate a desired therapeutic effect, such as pain relief. The precise desired therapeutic effect (e.g., the degree of pain relief, and source of the pain relieved, etc.) will vary according to the condition to be treated, the tolerance of the subject, the drug and/or drug formulation to be administered (e.g., the potency of the therapeutic agent (drug), the concentration of drug in the formulation, and the like), and a variety of other factors that are appreciated by those of ordinary skill in the art.
[00100] The term "transmucosal" delivery of a drug and the like is meant to encompass all forms of delivery across or through a mucosal membrane.

III. Drug Dosage Forms.
[00101] The claimed small volume oral transmucosal drug dosage forms produce a reduced saliva response as compared with conventional, larger dosage forms that are intended to deliver a drug in the oral cavity.
[00102] The preferred site for oral transmucosal drug delivery is the sublingual area, although in certain embodiments it may be advantageous for the dosage form to be placed inside the cheek, or to adhere to the roof of the mouth or the gum.
[00103] The dosage forms provide for the delivery of a greater percentage (and amount) of the drug via the oral mucosa and a corresponding decrease in delivery via the gastrointestinal (GI) tract as compared to traditional oral dosage forms and other oral transmucosal dosage forms.
[00104] Typically, the dosage forms are adapted to adhere to the oral mucosa (i.e.
are bioadhesive) during the period of drug delivery, and until most or all of the drug has been delivered from the dosage form to the oral mucosa.
[00105] More specifically, the dosage forms have a mass of less than 100mg, 90mg, 80mg, 170mg, 60mg, 50mg, 40mg, 30mg, 29mg,28mg, 27mg, 26mg, 25mg, 24mg, 23mg, 22mg, 21mg, 20mg, 19mg,18mg, 17mg, 16mg, 15mg, 14mg, 13mg, 12mg, 11mg, 10mg, 9mg, 8mg, 7mg, 6mg or 5mg or a volume of less than 100p1, 90pl, 80pl, 70pl, 60 pl, 50pl, 40pl, 30pl, 29 pl, 28 pl, 27pl, 26pl, 25pl, 24pl, 23pl, 22pl, 21pl, 20pl, 19pl, 18pl, 17pl, 16pl, 15pl, 14pl, 13pl, 12pl, 11p1, 10p1, 9pl, 8pl, 7pl, 6pl or 5pl.
[00106] In a preferred embodiment, the claimed dosage forms have a mass of less than 30mg and a volume of less than 30u1.
[00107] The dosage forms typically have bioadhesive characteristics and may form a hydrogel upon contact with an aqueous solution.
[00108] The dosage forms typically have an erosion time of from about 6 minutes or up to 25 minutes, however the erosion time may vary. More specifically, the dosage forms typically have an erosion time of about 5 minutes, 6 minutes, 7 minutes, 8 minutes, 9 minutes, 10 minutes, 11 minutes, 12 minutes, 13 minutes, minutes, 15 minutes, 16 minutes, 17 minutes, 18 minutes, 19 minutes, 20 minutes, 21 minutes, 22 minutes, 23 minutes, 24 minutes or 25 minutes.
[00109] In general, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or at least 99% of the total amount of pharmaceutically active substance in a dosage form administered to the oral mucosa of a subject is absorbed via the oral transmucosal route.
[0110] The dosage forms may have essentially any shape, examples of which include a round disc with a flat, concave, or convex face, an ellipsoid shape, a spherical shape, a polygon with three or more edges and flat, concave, or convex faces. The dosage forms may be symmetrical or asymmetrical, and may have features or geometries that allow for controlled, convenient, and easy storage, handling, packaging or dosing.
[0111] Oral transmucosal drug delivery is simple, non-invasive, and can be accomplished by a caregiver or patient with minimal discomfort. A dosage form for oral transmucosal delivery may be solid or non-solid. In one preferred embodiment, the dosage from is a solid that turns into a hydrogel following contact with saliva. In another preferred embodiment, the dosage from is a solid that erodes without forming a hydrogel following contact with saliva.
[0112] Generally, oral transmucosal delivery of pharmaceutically active substances is achieved using solid dosage forms such as lozenges or tablets, however, liquids, sprays, gels, gums, powders, and films and the like may also be used.
[0113] For certain drugs, such as those with poor bioavailability via the GI
tract, e.g., lipophilic opioids such as sufentanil, oral transmucosal delivery is a more effective delivery route than GI delivery. For such lipophilic drugs, oral transmucosal delivery has a shorter onset time (i.e., the time from administration to therapeutic effect) than does oral GI delivery and provides better bioavailability and more consistent pharmacokinetics.
[0114] The small size of the claimed drug dosage forms is designed to reduce the saliva response, thus reducing the amount of drug swallowed, and thereby delivering a substantial amount of drug to a subject via the oral mucosa. The claimed drug dosage forms provide for efficacious delivery of sufentanil via the oral mucosa and a consistent plasma level within the therapeutic window.
[0115] Formulations for preparation of the claimed dosage forms and methods of making them are described in US Application Serial Nos. 11/825,251 and 11/650,227.
An exemplary formulation is bioadhesive and comprises from about 0.0004% to about 0.04% sufentanil, e.g., 0.0005%, 0.001%, 0.002%, 0.003%, 0.004%, 0.006%, 0.008%, 0.01%, 0.012%, 0.014% or 0.016% sufentanil. In general, the formulation comprises(a) a non-ordered mixture of a pharmaceutically active amount of a drug; (b) a bioadhesive material which provides for adherence to the oral mucosa of the subject; and (c) stearic acid, wherein dissolution of a dosage form comprising the formulation is independent of pH, e.g., over a pH range of about 4 to 8.
[0116] Pharmaceutically active agents for use in a formulation of the invention are characterized by logarithm of the octanol-water partition coefficient (LogP) between 0.006 and 3.382.
[0117] A pharmaceutical dosage form of the invention for oral transmucosal delivery may be solid or non-solid. In one preferred embodiment, the dosage from is a solid that transforms into a hydrogel following contact with saliva. In another preferred embodiment, the dosage form is a solid that transforms into a bioadhesive film upon contact with saliva.
[0118] In a preferred embodiment, such formulations are designed to form a film which is visible following tablet disintegration. Upon placement on the oral mucosa, the dosage form absorbs water such that upon full hydration, it spreads across the mucosal surface, thus transforming into a bioadhesive film containing the active drug. This transformation results in a significant increase of the surface area available for drug release, thus accelerating drug diffusion and release from the dosage form. Owing to the higher contact surface area, drug absorption occurs fast, resulting in fast onset of action.
[0119] Numerous suitable nontoxic pharmaceutically acceptable carriers for use in oral dosage forms can be found in Remington's Pharmaceutical Sciences, 17th Edition, 1985.
[0120] It will be understood that the formulation is converted into a dosage form for delivery to a subject using procedures routinely employed by those of skill in the art, such as direct compression, wet granulation, etc. The process for preparation of the dosage form is optimized for each formulation in order to achieve high dose content uniformity.
[0121] While not wishing to be bound by theory, in one exemplary embodiment, when a drug dosage form is placed in the sublingual cavity, preferably under the tongue on either side of the frenulum linguae, it adheres upon contact. As the dosage form is exposed to the moisture of the sublingual space the dosage form absorbs water, resulting in erosion of the dosage form and release of the drug to the circulation of the subject.

IV. Sufentanil.
[00122] Opioids are widely used for the treatment of pain, and are generally delivered intravenously, orally, epidurally, transdermally, rectally and intramuscularly.
Morphine and its analogues are commonly delivered intravenously and are effective against severe, chronic and acute pain. However, they can also have severe respiratory depressive effects if not used appropriately and also suffer from a high abuse potential. The predominant cause of morbidity and mortality from pure opioid overdoses is due to respiratory complications.
[00123] Sufentanil (N-[(4-(Methoxymethyl-l-(2-(2-thienyl)ethyl)-4-piperidinyl)]-N-phenylpropanamide), is used as a primary anesthetic, to produce balanced general anesthesia in cardiac surgery, for epidural administration during labor and delivery and has been administered experimentally in both intranasal and liquid oral formulations. A commercial form of sufentanil used for IV delivery is the SUFENTA
FORTE formulation. This liquid formulation contains 0.075 mg/mi sufentanil citrate (equivalent to 0.05 mg of sufentanil base) and 9.0 mg/mi sodium chloride in water.
It has a plasma elimination half-life of 148 minutes, and 80% of the administered dose is excreted in 24 hours.
[00124] The use of sufentanil clinically has predominantly been limited to IV
administration in operating rooms or intensive care units. There have been a few studies on the use of liquid sufentanil preparations for low-dose intranasal administration (Helmers et al., 1989; Jackson K, et al., J Pain Symptom Management 2002: 23(6): 450-452) and case reports of sublingual delivery of a liquid sufentanil preparation (Gardner-Nix J., J Pain Symptom Management. 2001 Aug; 22(2):627-30;
Kunz KM, Theisen JA, Schroeder ME, Journal of Pain and Symptom Management, 8:189-190, 1993). In most of these studies, the smallest dosing of sufentanil in adults was 5mcg in opioid naive patients. Liquid administered to the oral or nasal mucosa suffers from lower bioavailability and possibly a shorter duration of action as demonstrated by the animal studies (sublingual liquid) described herein, as well as the literature (nasal liquid drops - Helmers et al., 1989). Gardner-Nix provides analgesic data (not pharmacokinetic data) produced by liquid sublingual sufentanil and describes the analgesic onset of liquid sublingual sufentanil occurring within 6 minutes but the duration of pain relief lasted only approximately 30 minutes.
[00125] A number of opioid dosage forms many of which contain fentanyl are currently available for treatment of pain.
[00126] Following transbuccal administration of fentanyl using a lozenge (e.g., Actiq ), the bioavailability is 50%, although the Tmax for the 200 mcg dosage of Actiq ranges from 20 - 120 minutes resulting from erratic GI uptake due to the fact that 75% of the fentanyl is swallowed (Actiq package insert). More recent publications on the Tmax of Actiq indicate that these original times were skewed towards more rapid onset (Fentora package insert indicates a range of Tmax for Actiq extending up to 240 minutes). Fentora (a fentanyl buccal tablet) exhibits a bioavailability of 65%, with reported swallowing of 50% of the drug. In contrast to the claimed dosage forms, both Actiq and Fentora suffer from the disadvantage that substantial amounts of lozenge-administered fentanyl are swallowed by the patient.
[00127] Sufentanil and fentanyl have many similarities as potent mu-opioid receptor agonists, however, they have been shown to differ in many key ways.
Multiple studies have demonstrated sufentanil to be in the range of 7 - 24 times more potent than fentanyl (SUFENTA package insert; Paix A, et al. Pain, 63:263-69, 1995; Reynolds L, et al., Pain, 110:182-188, 2004). Therefore, sufentanil may be administered using a smaller dosage form, avoiding the increased saliva response of a larger dosage form and thereby minimizing the amount of drug that is swallowed.
This leads to minimal GI uptake.
[00128] In addition, fentanyl and other opiate agonists, have the potential for deleterious side effects including respiratory depression, nausea, vomiting and constipation.
[00129] There is evidence which suggests that sufentanil may have less respiratory depression than fentanyl and other opioids at clinical doses (Ved et al., 1989; Bailey et al., 1990; Conti et al., 2004).
[00130] Since fentanyl has a 30% bioavailability from the GI route, swallowed drug can contribute to the Cmax plasma levels to a significant degree and results in the erratic Cmax and Tmax observed with these products. In contrast, the bioavailability of sufentanil from the GI route is 10-12%, and therefore swallowed drug will not contribute to the Cmax plasma levels to a significant degree.
[00131] Further, the lipid solubility (octanol-water partition coefficient) of sufentanil (1778:1) is greater than fentanyl (816:1) (van den Hoogen and Colpaert, Anesthes. 66:186-194,1987). Sufentanil also displays increased protein binding (91 -93%) compared with fentanyl (80 - 85%) (SUFENTA and Actiq package inserts, respectively). Sufentanil has a pKa of 8.01, whereas the pKa of fentanyl is 8.43 (Paradis et al., Therapeutic Drug Monitoring, 24:768-74, 2002). These differences can affect various pharmacokinetic parameters, for example, sufentanil has been shown to have a faster onset of action and faster recovery time than fentanyl (Sanford et al., Anesthesia and Analgesia, 65:259-66, 1986). As compared to fentanyl, use of sufentanil can result in more rapid pain relief with the ability to titrate the effect and avoid overdosing.
[00132] Importantly, sufentanil has been shown to produce endocytosis of the mu-opioid receptor 80,000 times more potently than fentanyl (Koch et al., Molecular Pharmacology, 67:280-87, 2005). The result of this receptor internalization is that neurons continue to respond to sufentanil more robustly over time than with fentanyl, suggesting that clinically less tolerance would develop to sufentanil compared to fentanyl with repeated dosing.
[00133] Prior to the work of the current inventors, no pharmacokinetic data had been published on sublingual sufentanil in any form. Pharmacokinetic data for ocular and intranasal transmucosal delivery of sufentanil has been published based on studies in dogs and humans. Farnsworth et al. (Anesth Analg, 1998, 86:138-140) describe ocular transmucosal absorption and toxicity of sufentanil in dogs, where 50 mcg of sufentanil was administered over a period of 2.5 minutes to the conjuctiva of five anesthetized dogs. The Tmax occurred at 5 min with a Cmax of 0.81 ng/mL
and a t1/Z of approximately 18 minutes. A study report of intranasal and intravenous administration of 15mcg of sufentanil in 16 humans provides a comparison of pharmacokinetic profiles, where intranasal sufentanil was delivered via 3 drops in each nostril with 2.5 mcg/drop. Intranasal sufentanil had a 78%
bioavailability based on the AUC from 0 - 120 minutes compared with intravenous delivery.
Intranasal delivery resulted in a Tmax of 10 minutes with a Cmax of 0.08 ng/mL. The t1/Z was approximately 80 minutes. See, Helmers et al., Can J Anaesth. 6:494-497, 1989. A third study in pediatric patients describes preoperative intranasal dosing of 15 children with 2 mcg/kg sufentanil via nasal drops and with plasma levels of sufentanil measured starting at 15 minutes, which was too late to capture the Tmax.
Based on extrapolation of the data, the Cmax was approximately 0.3 ng/mL and the ti/Z was approximately 75 minutes (Haynes et al., Can J Anaesth. 40(3):286, 1993).
Sufentanil Dosaae Forms
[00134] The active agent in the claimed dosage forms is sufentanil, alone or in combination with a sufentanil congener such as alfentanil, fentanyl, lofentanil, carfentanil, remifentanil, trefentanil, or mirfentanil. In a preferred embodiment, sufentanil alone is the active agent. Sufentanil may be provided in the claimed dosage forms in any of a number of formulations. Sufentanil may be provided as sufentanil citrate, sufentanil base, or a combination thereof.
[00135] A sufentanil drug dosage form may contain from about 0.25 to about 200mcg of sufentanil per dosage form for sublingual delivery. In one exemplary embodiment, each dosage form contains from about 0.25 to about 200mcg of sufentanil, alone or combination with one or more other therapeutic agents or drugs.
[00136] Exemplary drug dosage forms for administration to children (pediatric patients) contain from about 0.25 to about 120mcg of sufentanil per dosage form.
For example, a drug dosage form for administration to children may contain about 0.25, 0.5, 1, 2.5, 4, 5, 6, 8, 10, 15, 20, 40, 60 or 120mcg of sufentanil for oral transmucosal delivery. It follows that for pediatric patients, an exemplary dose range is from at least about 0.02 mcg/kg to about 0.5 mcg/kg with a preferable range of from about 0.05 to about 0.3 mcg/kg.
[00137] Exemplary drug dosage forms for administration to adults contain from about 2.5 to about 200mcg of sufentanil per dosage form. For example, a drug dosage form for administration to adults may contain about 2.5, 3, 5, 7.5, 10, 15, 20, 40, 60, 80, 100, 120, 140, 180 or 200mcg or more of sufentanil for oral transmucosal delivery.
[00138] Preferably, a sufentanil-containing dosage form comprises from about 5 to about 100 micrograms (mcg) of sufentanil, e.g., 5 mcg, 10 mcg, 15 mcg, 20mcg, 30mcg, 40mcg, 50mcg, 60mcg, 70mcg, 80 mcg or 100mcg of sufentanil.
[00139] As will be understood by those of skill in the art, the dose will be on the low end of the range for children and the high end of the range for adults dependent upon body mass, in particular when administered long term to opioid-tolerant adults.
Prior to the work of the current inventors, small-volume sufentanil-containing dosage forms for oral transmucosal drug delivery had not been described.
[00140] In various embodiments, the claimed dosage forms provide effective pain relief in all types of patients including children, adults of all ages who are opioid tolerant or naive and non-human mammals. The invention finds utility in both the inpatient and outpatient setting and in the field.
Conaeners of Sufentanil
[00141] Congeners of sufentanil find use in the compositions, methods and systems described herein, examples of which include alfentanil, lofentanil, carfentanil, remifentanil, trefentanil or mirfentanil.
[00142] In certain embodiments, the dosage form comprises at least 0.005% to as much as 99.9% by weight of alfentanil, lofentanil, carfentanil, remifentanil, trefentanil or mirfentanil. The percentage of active ingredient(s) will vary dependent upon the size of the dosage form and nature of the active ingredient(s), optimized to obtain maximal delivery via the oral mucosal route. In some aspects of the invention, more than one active ingredient may be included in a single dosage form.

V. Treatment Of Pain.
[00143] Using current treatment methods, pain control is attempted using a number of interventions, which generally include: patient-controlled analgesia (PCA), continuous epidural infusion (CEI), other types of acute pain control, palliative care pain control, and home health patient pain control. These methods meet with varying degrees of success with respect to duration of control, ease of treatment and safety versus side effects.
[00144] The need for rapid treatment of acute pain occurs in many different clinical situations, including post-operative recuperation, rheumatoid arthritis, failed back, end-stage cancer (i.e., breakthrough pain), etc. Post-operatively, for example, patients suffer from severe pain for the first few days followed by days of mild to moderate levels of pain.
[00145] The most common analgesic used to treat moderate to severe post-operative pain is IV morphine. This is either delivered on an "as needed"
basis to the patient by a nurse using IV injection or commonly a morphine syringe is placed in a PCA pump and the patient self-administers the opioid by pressing a button which has a lock-out feature. Other opioids, such as hydromorphone and fentanyl may also be administered in this manner.
[00146] Treatment of acute pain is also necessary for patients in an outpatient setting. For example, many patients suffer from chronic pain and require the use of opioids on a weekly or daily basis to treat their pain. While they may have a long-acting oral or transdermal opioid preparations to treat their chronic underlying pain levels, they often need short-acting potent opioids to treat their severe breakthrough pain levels.
[00147] Treatment of acute pain is also necessary "in the field" under highly sub-optimal conditions. Paramedics or military medics often are required to treat severe acute pain in un-sterile situations, where needles used for IV or IM
administration can result in unintended needle sticks, risk of infection, etc. Oral opioid tablets often take 60 minutes to provide relief which is too long for someone in severe pain.
[00148] In a number of clinical settings, there is clearly a need for improved means to administer a drug that produces effective pain relief in a manner that is titratable, safe and convenient, and non-invasive that provides relief from acute, severe breakthrough or intermittent pain over an appropriate period of time.
[00149] The claimed compositions, methods and systems rely on administration of dosage forms comprising a pharmaceutically active substance such as sufentanil which is effective for the treatment of acute pain (i.e. post-operative pain), intermittent pain or breakthrough pain, using a dispensing device that includes features such as lock-out, a means for user identification prior to drug administration and a means to protect the dosage forms stored in the dispensing device. The claimed methods and systems thereby provide significant advantages over currently available treatment modalities in terms of both safety and efficacy.

VI. In Vivo Human studies
[00150] Provided herein is pharmacokinetic data obtained in humans based on studies where sufentanil was administered via the sublingual route using the claimed small volume dosage forms.
[00151] Two human clinical studies were performed using healthy human volunteers. The first study which is detailed in Example 1 was performed with subjects (6 men and 6 women) using slow-eroding sublingual sufentanil dosage forms containing either 2.5mcg, 5mcg or 10mcg of sufentanil base corresponding to 3.7mcg, 7.5mcg or 15 mcg of sufentanil citrate, respectively in comparison to a 10-minute IV infusion of 5mcg sufentanil or 4 repeated doses of a slow-eroding sublingual sufentanil dosage form containing 5mcg sufentanil administered at 10-minute intervals (Table 1). The second study which is detailed in Example 2 was performed with 11 subjects using faster-eroding sublingual sufentanil dosage forms containing either 10mcg or 80mcg of sufentanil base corresponding to 15mcg or 120mcg of sufentanil citrate, respectively, in comparison to a 10-minute IV
infusion of 10mcg sufentanil or a 20-minute IV infusion of 50mcg sufentanil, a sublingual dose of 5mcg of sufentanil solution or 4 repeated administrations of fast-eroding sublingual sufentanil dosage forms containing 10mcg of sufentanil administered at 20-minute intervals (Table 2). All excipients were "pharmaceutically acceptable"
(inactive) and have GRAS or "generally recognized as safe" status.
[00152] Sufentanil dosage forms designed for sublingual use were compared to IV
sufentanil, administered through an IV catheter as a continuous infusion.
Plasma samples were drawn from a different IV catheter at a remote location. The assay demonstrated good inter-day precision and accuracy at the high, medium and low quality control sample concentrations.
[00153] The dosage forms for the first study eroded over a period of 15-25 minutes in all subjects and are designated herein as "slow-eroding". The dosage forms for the second study eroded over a period of 6-12 minutes in all subjects and are designated herein as "faster-eroding". After placement of each sufentanil dosage form in the sublingual cavity of the healthy volunteers, a remarkably consistent pharmacokinetic profile was obtained. The bioavailability of sufentanil administered using small volume sublingual dosage forms as compared to IV administration for single and multiple administrations was high and ranged from 60.9% (10mcg dose;
faster-eroding) to 97.2% (4 x 5mcg dose (slow-eroding). The bioavailability of sufentanil administered using small volume sublingual dosage forms is greater than that of the fentanyl products, Actiq and Fentora (47% and 65%, respectively -Fentora package insert). Importantly, the bioavailability is linked to the consistency of total drug delivered to the patient. For example, the total plasma drug area under the curve (AUC 0-infinity) for sufentanil dosage forms 10mcg was 0.0705 +
0.0194 hr*ng/ml (mean + standard deviation (SD)).This SD is only 27.5% of the total AUC.
Coefficient of variation (CV) is a term to describe the percent SD of the mean. The coefficient of variation for the fentanyl products, Fentora (AUC is 45%) and Actiq (AUC is 41%; Fentora package insert), while the coefficient of variation around the bioavailability of sufentanil administered using small volume sublingual dosage forms is less than 40%. Therefore, the total dose delivered to the subject is not only more bioavailable for the sufentanil dosage forms but it is more consistent.
[00154] Although this high bioavailability could be due to a number of factors including but not limited to erosion time, it is likely that the lack of saliva produced by the small size of the dosage forms limits the swallowing of the drug and avoids the low bioavailability typical of drug absorption via the GI route. Both Fentora and Actiq package inserts claim at least 50% and 75% of the drug dose, respectively, is swallowed via the saliva, and both exhibit lower bioavailability than the claimed dosage forms.
[00155] The dosage forms used in the clinical trials had a volume of approximately 5 microliters (mass of 5.5-5.85 mg), a small fraction of the size of Actiq or Fentora lozenges. Therefore, less than 25% of the drug is swallowed, which is a much lower percentage than is swallowed with Fentora or Actiq.
[00156] The sufentanil sublingual dosage forms are also superior in terms of consistent drug plasma levels early after administration. The Cmax obtained with the 10 mcg sufentanil dosage form was 27.5 + 7.7 pg/ml. The coefficient of variation of the Cmax is therefore only 28%. The Cmax for Fentora and Actiq suffer from variability of GI uptake of drug. Fentora reports a Cmax of 1.02 0.42 ng/ml, therefore the coefficient of variation of the Cmax is 41%. The range of coefficients of variation for the various doses of Fentora is from 41% to 56% (package insert). The Actiq coefficient of variation of Cmax is reported as 33% (Fentora package insert).
[00157] In addition to superior bioavailability and consistency in plasma concentrations, the Tmax for 10 mcg sufentanil dosage forms was 40.8 13.2 minutes (range 19.8 - 60 minutes). The reported average Tmax for Fentora is 46.8 with a range of 20 - 240 minutes. The Tmax for Actiq is 90.8 minutes, range 35 minutes (Fentora package insert). Therefore, the consistency in onset of analgesia for sufentanil dosage forms is markedly better than that of Fentora and Actiq.
[00158] In addition, the Tmax values obtained following repeated sublingual administration of the claimed sufentanil dosage forms were significantly shorter than those observed following administration of a single sublingual sufentanil dosage form. Most notably, the Tmax obtained with repeat dosing of 10 pg (4 x 10 pg) sufentanil dosage forms (fast-eroding) occurred 24.6 minutes after the previous (fourth) dose. The coefficient of variation around Tmax was only 18%, indicating a very consistent and predictable Tmax with repeated sublingual administration of the claimed sufentanil dosage forms.
[00159] The linearity of sufentanil plasma levels following sublingual administration of the claimed sufentanil dosage forms doses was consistent from the 2.5mcg dose up through the 80mcg dose.
[00160] Although still in development, published data allows comparison of the sufentanil pharmacokinetic data provided herein to that of Rapinyl, a fentanyl sublingual fast-dissolve lozenge. The coefficient of variation around the AUC
for all three doses of sufentanil exemplified herein (2, 5, and 10 mcg) averaged 28.6%, demonstrating that the observed low coefficient of variation is not dependent on dose. In contrast, the published bioavailability for a sublingual fentanyl product, Rapinyl, is approximately 70% (Bredenberg, New Concepts in Administration of Drugs in Tablet Form, Acta Universitatis Upsaliensis, Uppsala, 2003). The coefficient of variation of the AUC (0-infinity) for Rapinyl ranges from 25-42% and is dose-dependent.
[00161] In addition, the coefficient of variation of the Cmax for Rapinyl varies from 34 - 58% depending on dose. As shown by the data presented herein, administration of the 10 mcg sufentanil dosage form resulted in a Cmax with a coefficient of variation of only 28%, and the average coefficient of variation of Cmax for the 2, 5, and 10 mcg doses was 29.4%, indicating minimal variability depending on dose. Similarly, the coefficient of variation for Tmax with Rapinyl ranges from 43 -54% depending on dose, whereas for our sufentanil dosage forms, this coefficient of variation for Tmax averages only 29% over all three dosage strengths. This consistent onset of action achieved with sublingual sufentanil dosage forms allows a safer redosing window when compared to any of the three comparator drugs, since rising plasma levels are contained to a shorter period.
[00162] Additionally, as with Fentora and Actiq, Rapinyl demonstrates a longer plasma elimination half-life (5.4 - 6.3 hours, depending on dose) than the claimed sufentanil dosage forms. The plasma elimination half-life of sufentanil dosage forms ranged from 1.5 - 2 hours following a single oral transmucosal administration in humans (Table 2), which allows for more titratability and avoids overdosing.
As will be understood by those of skill in the art, the half-life described herein for the exemplified dosage forms may be adjusted by modification of the component and relative amounts of the excipients in the formulation used to make a given dosage form. The ability to titrate to higher plasma levels by administering repetitive doses of the sublingual sufentanil dosage forms was also tested in this human study.
[00163] The methods and systems described herein are designed to work effectively in the unique environment of the oral cavity, providing for higher levels of drug absorption and pain relief than currently available systems. The claimed methods and systems are designed to avoid the high peak plasma levels of intravenous administration by entry into the circulation via the sublingual mucosa.
[00164] The claimed methods and systems further provide for independent control of bioadhesion, dosage form disintegration (erosion) and drug release over time, together with administration using a device to provide a safe delivery profile. The device-administered sublingual dosage forms provide individual, repetitive doses that include a defined amount of the active agent (e.g., sufentanil), thereby allowing the patient or care giver to accurately titrate the amount of drug delivered and to adjust the amount as appropriate in a safe and effective manner. The lock-out feature of the dispensing device adds to the safety of the drug delivery profile.
[00165] Further, treatment with the claimed compositions, methods and systems provides for improved safety by minimizing the potentially deleterious side effects of the peaks and troughs in the plasma drug pharmacokinetics, which are typical of currently available medications or systems for treatment of pain.
[00166] Advantages of the claimed sublingual dosage forms over various liquid forms for either sublingual or intranasal administration include local release of drug from the dosage form over time with minimal swallowing of liquid drug via either the nasal or oral/GI route.
[00167] Due to the small size of the oral transmucosal dosage forms, repeated placement in the sublingual cavity over time is possible. Minimal saliva production and minimal physical discomfort occurs due to the small size, which allows for repetitive dosing over days to weeks to months. Given the lipid profile of the sublingual cavity, the sublingual route, also allows for slower release into the plasma for certain drugs, such as sufentanil, which may be due to utilization of a "depot"
effect that further stabilizes plasma levels compared to buccal delivery.
[00168] The oral transmucosal dosage forms are designed to fit comfortably under the tongue such that the drug form erodes sufficiently slowly to avoid the immediate peak plasma levels followed by significant drop off seen in prior art formulations such as described in US. Patent No. 6,759,059 (Rapinyl), wherein fentanyl was administered via tablets containing 400mcg of fentanyl which resulted in a peak plasma level of 2.5ng/ml followed by an immediate drop in plasma level.
Fentora (fentanyl buccal tablet) also suffers from a lack of a plateau phase but rather has a steep incline up to the Cmax followed by a significant drop-off in plasma levels (Fentora package insert).

VII. Utility Of Small-Volume Oral Transmucosal Dosaae Forms.
[00169] The claimed dosage forms, methods and systems find utility in delivery of sufentanil via the oral transmucosal, e.g., sublingual, route for treatment of pain.
The small volume oral transmucosal dosage forms provide for high bioavailability, low variability in Tmax, low variability in Cmax and low variability in AUC.
The dosage forms also provide for prolonged plasma levels within the therapeutic window.
[00170] More specifically, the claimed dosage forms, methods and systems provide the advantages that:

(a) there is a linear relationship between sufentanil plasma levels in a subject following administration of the claimed sufentanil dosage forms and the amount of sufentanil in the dosage form;
(b) a single sublingual administration of the claimed sufentanil dosage forms to a subject results in an AUC;nf with a coefficient of variation of less than 40%;
(c) a single or repeated sublingual administration of the claimed sufentanil dosage forms to a subject results in a Tmax with a coefficient of variation of less than 40%;
(d) repeated sublingual administration of the claimed sufentanil dosage forms to a subject results in a bioavailability that is greater than the bioavailability following a single sublingual administration to said subject;
(e) the difference between the Tmax following repeated sublingual administration of the claimed sufentanil dosage forms and the time of the previous sublingual administration is shorter than the Tmax following a single sublingual administration to the subject;
(f) there is a linear relationship between Cmax and the amount of sufentanil in the dosage form;
(g) there is a linear relationship between AUC;nf and the amount of sufentanil in the dosage form; and (h) the highest predicted steady-state sufentanil concentration following multiple administrations of 10 or 15mcg sublingual sufentanil dosage forms is predictable allowing for an accurate determination of safe lock-out times and therefore safe and efficacious treatment of pain.
[00171] In one exemplary embodiment described in detail herein, the dosage forms find utility in treating a subject suffering from pain that may be associated with any of a variety of identifiable or unidentifiable etiologies. In this embodiment, the dosage forms find utility in suppression or mitigation of pain. The term "treatment" or "management" of pain is used here to generally describe regression, suppression, or mitigation of pain so as to make the subject more comfortable, as determined for example by pain score.
[00172] The invention finds utility in the treatment of both opioid naive patients and opioid tolerant patients.
[00173] The dosage forms find particular utility in the treatment of acute pain, such as post-operative pain, as well as other pain, such as "in the field", i.e., under highly sub-optimal conditions.
[00174] Paramedics or military medics often are required to treat severe acute pain or other injuries or conditions in non-sterile situations, where needles used for IV or IM administration can result in unintended needle sticks, risk of infection, etc. Oral opioid tablets often take 60 minutes to provide relief which is too long for someone in severe pain. The claimed dosage forms find utility in addressing this need.
[00175] When the dosage forms are used for the treatment of pain, the claimed methods and systems find utility in administration of drugs to pediatric and adult populations and in treatment of human and non-human mammals, as well as in opioid tolerant and opioid naive patient populations.
[00176] Application of the claimed methods and systems is not limited to any particular therapeutic indication. As such, the claimed dosage forms find utility in administration of sufentanil to pediatric and adult subjects and in the treatment of human and non-human mammals.
[00177] The dosage forms find utility in pediatric applications, since the comfortable and secure nature of the dosage form allows children to readily accept this mode of therapy and will reliably deliver drug transmucosally. Specific examples include, but are not limited to, treatment of pediatric acute pain when IV
access is not available or inconvenient, treatment of pediatric asthma when the child is not able to use an inhaled route of administration effectively, treatment of nausea when a child can not or will not swallow a pill, pre-procedural sedation when a child is NPO
(no oral intake allowed) or a more rapid onset is required.
[00178] The dosage forms find further utility in veterinary applications.
Specific examples include, but are not limited to, any treatment of an acute condition for which IV administration is not readily available or inconvenient, such as pain relief, anxiety/stress relief, pre-procedural sedation, etc.
VIII. Dispensing Devices.
[00179] Dispensing devices and systems for oral transmucosal administration of small volume drug dosage forms are provided. The dispensing devices are handheld and portable and comprise a housing having a dispensing end which typically has a proboscis with a shroud that provide a means for blocking or retarding saliva ingress and/or moisture control. The dispensing devices further provide safety features such as a means for lock-out and a means for user identification.
[00180] The claimed dispensing devices, methods and systems comprise delivery of small volume dosage forms to the oral mucosa. The invention is not limited to the specific devices, systems, methodology and dosage forms detailed herein, as these may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention.
Blocking/Retarding Saliva and Moisture Ingress
[00181] In some embodiments, the claimed dispensing devices comprise a means for minimizing or eliminating saliva ingress and moisture ingress into the dispensing device: (1) to avoid wetting the dosage forms therein; (2) to isolate any saliva that enters the dispensing device in such a manner that the dosage forms therein remain dry; (3) to absorb or adsorb any saliva that enters the dispensing device in such a manner that the dosage forms remain dry; (4) to block saliva and moisture from entering the device to protect the dosage forms from vapor and liquid phase moisture, or (5) any combination thereof.
[00182] The dispensing device may have a means for preventing and/or controlling humidity ingress due to ambient conditions outside of the device.
[00183] The means for minimizing or eliminating saliva ingress or preventing other moisture from entering the dispensing device includes, but is not limited to, one or more flexible or rigid seals, one or more flexible or rigid wipers, use of one or more absorbent material components such as a desiccant or pad, a door or latch that is manually or automatically opened and closed, multiple stage delivery systems, a positive air pressure and airflow, or an air gap or prescribed distance or barrier/shroud maintained between the dosage form delivery orifice and the mucous membrane tissues within the mouth that may transport the saliva. The shroud limits the ability of the tongue or oral mucosa to contact the dosage form dispensing area, thereby controlling saliva contact and ingress. By inhibiting or eliminating the "wetness" inside the shroud and on the surface of the valve/seal, the dosage form is dispensed without adhesion occurring between the dosage form and the shroud or valve/seal.
[00184] To protect the drug dosage forms from exposure to moisture either from humidity, saliva ingress, or accidental exposure to other water based liquids, the dispensing device and the container or cartridge which houses the dosage forms within the device contains a desiccant.
[00185] Means for trapping or otherwise isolating saliva or moisture if it enters the device include, but are not limited to, a hydrophilic wicking material or component, an absorbent or adsorbent material or component, a desiccant material or component, a separate track or channel for moisture to collect, a separate channel to communicate moisture to the absorbents or adsorbents, or any combination of these materials or components.
[00186] A desiccant is a sorbant, in the form of a solid, liquid, or gel that has an affinity for water, and absorbs or adsorbs moisture from it's surrounding, thus controlling the moisture in the immediate environment. Any commercial desiccant may be used. Commercial desiccants typically take the form of pellets, canisters, packets, capsules, powders, solid materials, papers, boards, tablets, adhesive patches, and films, and can be formed for specific applications, including injection moldable plastics. There are many types of solid desiccants, including silica gel (sodium silicate, which is a solid, not a gel), alumino-silicate, activated alumina, zeolite, molecular sieves, montmorillonite clay, calcium oxide and calcium sulfate, or others, any of which may be used in the claimed dispensing devices. Different desiccants have different affinities to moisture or other substances, as well as different capacities, and rates of absorption or adsorption. Also, different types of desiccants will come to equilibrium at different relative humidities in their immediate surroundings. As a means for protecting the dosage forms and the internal portions of the dispensing device from moisture, one or more desiccants may be employed at the proboscis; in or adjacent to the dosage form; in or adjacent the delivery pathway; in or adjacent the dosage form, tablet magazine or cartridge; in or adjacent to other components of the dispensing device; formed as an injection molded component of the dispensing device; a compressed desiccant that is pressed into location; or desiccant in any other location within or without the device.
[00187] In one preferred embodiment, the desiccant snaps into a cavity in the side of the cartridge. There are holes in the desiccant cavity that connect it to the dosage form stack, exposing the dosage forms to desiccant and keeping them dry.
[00188] The claimed dispensing devices rely on valves, pads, seals, the rest position of the push rod, proboscis design and a shroud to minimize or eliminate saliva ingress or moisture into the dispensing device during administration of the dosage form.
[00189] Valves for use in the claimed devices are typically dome/trocar type valves that provide enough sealing force to keep saliva and/or moisture from entering the device and serve to minimize or eliminate saliva ingress or moisture by closing the distal orifice during dispensing and after a dosage form has been dispensed.
[00190] Pads for use in the claimed devices have various geometries that aid in contacting or communicating with the pushrod in order to removed liquid from the push rod surface. Such pads typically contain hydrophilic properties and serve to minimize or eliminate saliva ingress or moisture ingress by transporting the liquid away from the track and push rod.
[00191] Seals and wipers for use in the claimed devices are designed to maintain a uniform seal around a drug dosage form and a pushrod during delivery and are characterized by flexible materials that impart a seal around the dosage form and pushrod and serve to minimize or eliminate saliva ingress or moisture by sealing and wiping the orifice and pushrod before, during, and after dispensing.
[00192] The rest position of the push rod in the claimed devices is characterized by positioning the pushrod in an intermediate location distal to the cartridge exit, and proximal to the distal dispensing orifice and serves to minimize or eliminate saliva ingress and moisture by allowing the pushrod to reside in a location that contains a desiccant, absorbents, or channel that dries the pushrod while at rest between dosage dispenses.
[00193] The proboscis design for use in the claimed devices is characterized by a distal device shape, typically an S-shape, that aids in use of the device and/or placement of the tip on the oral mucosa of the subject. The shape typically has curves, angles, and geometries such that it enables proper use of the device and placement of the dosage form on the oral mucosa of the subject, e.g., in the sublingual space.
[00194] The shroud of the claimed devices has a geometry that forms a barrier between the device and the oral mucosa and tongue, a relief for dosage form delivery, and an interior that is hydrophobic or hydrophilic and serves to minimize or eliminate saliva ingress or moisture ingress by creating a barrier from the oral mucosa contacting the valve area and dosage form, aiding in dosage form dispensing and discouraging dosage form adherence to the shroud. The shroud may have a rounded interior surface or other geometries to mitigate the dosage form adhering to the shroud. The shroud limits the ability of the tongue or oral mucosa to contact the dosage form dispensing area, thereby controlling saliva contact and ingress.
[00195] Figs. 11A-E provide schematic depictions of a variety of aspects of one embodiment of a drug dispensing device constructed to hold a plurality of dosage forms for oral transmucosal delivery. Fig. 11A is a schematic depiction of a fully assembled or single piece dispensing device 11 of the invention. In Fig. 11B, the dispensing device 11 includes a reusable head 13 and a disposable body 15; in Fig.
11C the dispensing device 11 further includes a cartridge 17 in Fig. 11D the dispensing device 11 includes a valve 33, a proboscis 31, a latch button 19, a power train coupling 25, a hub lock 21 and a dispense button 23; and Fig. 11E
is a schematic depiction of a reassembled and complete dispensing device 11.
[00196] Fig. 12 provides a schematic depiction of an exemplary dispensing device wherein the dispensing tip comprises a shroud 29 having a one or more of: a wiping/sealing valve 37, an absorbent pad 39, a drug drying chamber/moisture communication channel 43, desiccant in the channel 45, a cartridge 17 containing dosage forms 67 and desiccant in the cartridge 47.
[00197] Figs. 13A and 13B are schematic depictions of an exemplary geometry for the dispensing tip that prevents contact of one or more seals 33, 35 with the moist or wet surface of the oral mucosa via a shroud 29.
[00198] Figs. 14A - D are a schematic depiction of an exemplary proboscis 31 of a dispensing device 11 wherein the proboscis 31 comprises a shroud 29, a valve for dispensing a dosage form 67 and a cut-out/relief 55 for the dosage form 67 to be placed against the oral mucosa and not moved when the device 11 is withdrawn following dispensing.
[00199] A means for minimizing saliva ingress and moisture into the claimed devices is important for preservation of the integrity of dosage forms during storage, e.g., prior to an between oral transmucosal administrations.
[00200] The claimed dispensing devices may be used to administer a drug dosage form that is sensitive to moisture and/or humidity. In such cases, a drug dosage form cartridge serves to protect the drug dosage form from liquid and vapor phase moisture, including humidity, liquid moisture, saliva, mucus, etc. The cartridge may be cylindrical, disk-shaped, helical, rectilinear, non-ordered, or may take the form of any assemblage of drug dosage forms that allows the drug dispensing device to dispense them in a controlled manner. To prevent the unused drug dosage forms from absorbing moisture or otherwise becoming exposed to moisture prior to use, the cartridge may provide a means of sealing the drug dosage forms from exposure to moisture. This may accomplished by use of a cartridge that contains individually packaged drug dosage forms separated by a thin impermeable foil or impermeable material such that when one drug dosage form is dispensed from the cartridge, the seal protecting the remaining dosage forms remains unbroken. Alternatively, the dosage forms may be packaged in such a manner within the cartridge that two or more dosage forms are packaged together in each separate sealed compartment.
In some embodiments, all of the dosage forms in a cartridge may be packaged together in a foil sealed compartment.
[00201] A drug cartridge that houses small volume drug dosage forms within the dispensing device may afford a seal against moisture by means of a septum, an elastomeric seal or valve, a sliding, translating, hinged door or valve, or by means of sealing against another component of the drug dispensing device when loaded.
In this manner, a single re-sealable seal may be opened either independently or by means of the passage of a dosage out of the cartridge. Once the dosage form is delivered from the cartridge, the re-sealable seal on the cartridge may be re-sealed to prevent moisture or other contaminants from damaging the remaining drug dosage forms within the cartridge. The cartridge may further have a non-re-sealable seal that is broken when it is loaded into the drug dispensing device or upon delivery of the first dosage form from the cartridge.
[00202] In other embodiments, the cartridge contains a desiccant or other absorbent or adsorbent material to absorb or adsorb moisture that penetrates the cartridge either prior to use or during normal use. A cartridge for use in a claimed dispensing device may contain any combination of individually sealed dosage forms, multiply sealed dosage forms, re-sealable seals, non-re-sealable seals, desiccants, absorbents, or adsorbents. In one embodiment, a cartridge for use in the dispensing device in holds sufficient drug dosage forms for 1-5 days of treatment, e.g.

dosage forms or sufficient drug dosage forms to provide 48 to 72 hours of treatment.
Pushrod Design
[00203] Figs. 15A - D provide a series of flow diagrams for use of an exemplary dispensing device showing pusher logic, wherein Fig. 15A shows the LOAD
feature;
Fig. 15B shows the device calibration logic flow. Referring to Fig. 16, the pushrod 51 is advanced from position 65, picks up the shipping tablet 69 at position 63, and is further advanced to position 61. At position 61, the device senses the presence of the shipping tablet 69 and/or push rod 51. In doing so, the device is calibrated and knows the location of the shipping tablet 69 and/or end of the push rod 51 regardless of assembly tolerances, variations in push rod length and push rod end conditions. Following this calibration, the push rod 51 advances the shipping tablet 69 from position 61 to position 57 where the shipping tablet 69 is dispensed from the device. During this operation, the device is able to distinguish between a shipping tablet 69, a push rod 51, and a drug dosage form 67. This differentiation enables the device to confirm that a cartridge is unused because a shipping tablets is the first thing dispensed from a new cartridge during device setup. The feature that provides the means for differentiating between the shipping tablet, push rod, and dosage form 67 may be optical, physical, RF, electronic (resistive, capacitive, or other) or magnetic. The push rod 51 advance from position 65 and position 57 described above, could be continuous or intermittent and a physical stop at position 61 is not required. The push rod 51 then retracts from position 57 to position 59, placing the device 11 in the ready position with the push rod 51 under the remaining dosage forms 67. In this position, the push rod 51 keeps dosage forms 67 from inadvertently falling out of the device 11.
[00204] Fig. 15C shows the device dispense logic flow. referencing Fig. 16, following a dose command, the push rod 51 retracts from position 59 to position 65, allowing the dosage forms 67 to advance into the push rod track. The push rod 51 then advances from position 65, picks up a dosage form at position 63, and then dispenses the dosage forms 67 from the device at position 57. Between positions 63 and 57, the presence of a dosage form 67 is sensed/confirmed at position 61 by the position sensor. The push rod then retracts from position 57 to position 59, placing it in the ready position with the push rod 51 is under the remaining dosage forms 67. In this position, the push rod 51 is allowed to dry before the next dosage form 67 dispense, as well as keeps dosage forms 67 from inadvertently falling out of the device 11.
[00205] Fig. 15D shows the device disassemble logic flow. Following a "disassemble" command, the push rod 51 is moved to position 65. This allows for the removal of any remaining dosage forms 67 without push rod interference.
[00206] Fig. 16 is a schematic depiction of an exemplary dispensing device showing the stages of push rod/ dosage form interaction during device use. In Fig 16, the push rod 51, dosage forms 67, shipping tablet 69, spring 73 and position sensor 71 are shown. During use, the push rod 51 moves between positions 57, 59, 61, 63 and 65, also shown in Fig. 16 and further detailed in Figs. 15A-D.
Dosing Historv/Feedback
[00207] Further embodiments of the device include the ability to store historical use information and the ability to transmit such information. The device may be capable of unidirectional (downloading) or bidirectional information transfer.
For example, an exchange of information may be accomplished by downloading stored information to a computer through a physically wired interface, such as a USB
or any other communication connection. Alternatively, information may be communicated via a wireless system.
[00208] In another embodiment, the dispensing device has a dose counting feature that monitors and stores the history of drug usage. Such information may include historical use information, for example the number of dosages stored and dispensed, and the times of dispensing.
Calibration
[00209] The dispensing device may be capable of self-calibration of the dispense mechanism, or the device may be calibrated manually. This process may employ a shipping tablet with a feature or features that physically differentiate it from a drug dosage form or the push rod. These features may be designed so that device calibration precision is higher that that attainable using a dosage form or push rod.
The differentiating feature may be physical, optical, radio frequency (RF), electronic or magnetic.
User identification Feature
[00210] In one aspect, the dispensing device comprises a detecting means for user identification such as a fingerprint reader, an optical retinal reader, a voice recognition system, a face recognition system, a dental imprint recognition system, a visual recognition system, or a DNA reader. The dispensing device may employ one or more means to identify the user, enabling the system to determine if a dispensing request is being made in an authorized or unauthorized manner. It is important for effective delivery of many potential drugs and drug dosage forms to ensure that the dispensing device is not accidentally or intentionally used by an unauthorized individual to prevent accidental or intentional diversion of the drug. Such user identification systems may recognize one or more users, for example, in an inpatient hospital setting the dispensing device could be programmed to recognize the patient to whom it is prescribed, as well as authorized healthcare providers such as nurses and physicians. In an outpatient home setting, for example, the dispensing device may only respond to the patient to whom it is prescribed.
[00211] The dispensing device may employ any means of user identification, including fingerprint identification, RFID detection with the use of an active or passive RFID tag on bracelet, necklace, clip, belt, strap, adhesive patch, implant, or means of locating and affixing a tag, retina identification, DNA
identification, voice recognition, password or code entry, physical key, electronic or magnetic key, personal area network identification using the human body or clothing as a data or signal conduit, optical scanner or face recognition, sonic, subsonic or ultrasonic identification, or any other means of identifying an individual and verifying their identity.
[00212] One method of user identification is the use of a short distance ("near field") passive RFID tag attached to a bracelet, necklace, adhesive patch, clothing tag, orally mounted device, like an orthodontic retainer, belt, strap, some combination of these, or another location. When an RFID tag is used in the "near field", roughly defined as about 16% of the wavelength of the received signal, the tag behaves in the inductive mode of operation, coupling between the reader and tag antenna magnetically. The near field is characterized by at least two features:
first is a rapid decline in field strength with distance, and second is a strong directionality of the signal. In the near field, the signal strength falls off very rapidly, with a signal strength loss of approximately 60dB per decade in distance. For good inductive coupling between the transmitter antenna and the RFID tag antenna, the two antennas are oriented in parallel planes with the axes through the center of each antenna in close proximity. Strong signal strength (robust user identification) is provided when the device is very close to the RFID tag. At the same time, a very poor signal is provided when the device is further away from the tag, which helps prevent unauthorized use by someone other than the patient who attempts to use the device. It is preferable to operate in this near field region with good antenna alignment. Furthermore, it is preferable to operate with a very short distance of adequate signal strength for a positive identification, so that it is very difficult to receive a signal if the device is not in the proper orientation and proximity to the RFID tag. To attain a short distance and a proper alignment between antennas, the dispensing device may be designed so as to properly locate the RFID reader antenna, mounted in the dispensing device, adjacent to an RFID tag antenna, mounted, for example, on a wrist band or bracelet, or a clothing tag on the collar, or an adhesive patch on the hand, arm, cheek, neck, or elsewhere. Furthermore, an RFID tag antenna on a wrist band or bracelet may be held in proper alignment and location by means of a small adhesive patch that prevents the bracelet from moving or rotation on the wrist.
[00213] In another embodiment, the dispensing device employs a high frequency RFID reader for use in the inpatient (hospital, clinic, etc.) setting, operating on or near the 13.56MHz frequency band, and the patient is be fitted with a matching RFID tag and antenna on a disposable bracelet or wrist band, designed in such a way that if the bracelet or wrist band is removed the RFID tag, the antenna, or another component of the associated circuit will be damaged or destroyed, rendering the bracelet or wrist band non-functional. In one example, the range of the RFID
communication is short, between 0 inches and 10 inches preferably, more preferably between 0 and 5 inches, and most preferably between 0 and 3 inches, and may additionally be directional, allowing proper use by the intended patient to be easy and reliable, while at the same time making unauthorized use by another individual difficult, very difficult, or impossible.
Lock Out
[00214] The dispensing device provides for lock out, requiring the patient to communicate with the physician or other authorized care giver to unlock the device for the next fixed period. In this way the device and dock provide for safe drug administration due to greater physician oversight and care management.
[00215] The dispensing device provides a means for adjusting both the initial dose and subsequent doses, as well as the lock-out time. The initial dose and lock out time may subsequently be adjusted dependent upon patient response, duration of treatment and the like.
[00216] The initial timed lock-out period for a claimed dispensing device is typically from about 1 minute to about 60 minutes, from 3 minutes to 40 minutes or from minutes to 30 minutes, and in particular cases is set at any one minute interval from 1 to 60 minutes, e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 ,16, 17, 18, 19, 20, 21, 22, 23 ,24, 25, 26, 27, 28, 29, 30, 31, 32, 33 ,34, 35, 36, 37, 38, 39,40, 41, 42, 43 ,44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59 or 60 minutes.
[00217] In some cases, a dispensing device has a fixed lockout between doses and may exhibit a shutdown after a fixed period of time. In other cases, the lock-out time is a programmable lock-out time. The lock-out time may also be a fixed time lock-out interval, a predetermined lock-out interval, a predetermined variable lock-out interval, a lock-out interval determined by an algorithm or a variable lock-out interval communicated to the device from a remote computer or docking station.
Additional Features
[00218] A dispensing device may provide the ability to recognize a specific cartridge by a mechanical, optical (e.g. bar code), electronic (e.g.
microchip), magnetic, radio frequency, chemical, or other means of detecting and identifying the cartridge. In one exemplary embodiment, the drug-containing cartridge contains a physical keying detail on the cartridge that is physically detected by a sensor or switch or a series of sensors or switches in the dispensing device.
Furthermore, the dispensing device may communicate unidirectionally or bi-directionally with the cartridge to exchange information. Such information may include drug name, dosage strength, usage information, lockout period, manufacturing lot number, indications for use, side effects, drug interactions, date of manufacture, date of expiration, serial number, number of doses in the cartridge, or any other relevant information.
The dispensing device may be able to write, in addition to read, information to the cartridge, like date used, health care provider or other user identification, number of doses used, etc.
[00219] The dispensing device may provide mechanical protection for the dosage forms contained therein, preventing breakage, chipping, hydration etc., thereby allowing for dispensing of the undamaged dosage forms contained therein. This is of particular importance for small fragile and friable dosage forms.
[00220] The drug dispensing device may be powered by a battery, capacitor, fuel cell, or other power supply source, or may require no electrical power, but be manually activated.
[00221] In some embodiments, the dispensing device is capable of issuing alarms or other notifications when functional or safety issues arise. The alarm or other notification may trigger an alert on the dispensing device, on a dock or other peripheral device; on a computer or by means of a wired or wireless network, or may alert other remote devices. The alarm or notification may be audible, tactile, visual, or may employ other means of notifying one or more individuals.
Docking Station
[00222] In certain embodiments, the device includes a portable or fixed docking station that may query the device, reset it between dosing, lock it when not properly accessed, and control the dosing regimen. The drug dispensing device may communicate with a physician or care giver, via the dock, or by a wired or wireless communication means.
[00223] The dispensing device may employ one or more levels of interface for different types of authorized users, for example the patient, the nurse, the physician, pharmacist or other authorized medical or healthcare personnel. These different interfaces may include components such as keypads, buttons, graphical icons and instructions, lights, LED's, monochrome or color graphical or text displays, touch-screens, LCD's, sounds, tactile feedback, voice recognition interfaces, and other input and output devices and means. The activity, or mode, of the user interface may be determined by the mode of operation of the dispensing device, by a login or access activity by a user such as a password or code entry, by the connection or disconnection of the dispensing device from a dock, computer, or network, or by the detection of an authorized access key, such as a key, and/or RFID tag, or similar combination. Upon changing the interface mode, the functionality of the device may be changed, either activating, inactivating or changing the functionality of the various interface components described above. By allowing the device to have one or more interface modes, with differing functionality associated with each one, the device can be optimized for various uses.
Base Station In some embodiments the drug dispensing system includes a base station for recharging the drug dispensing device and the portable docking FOB between uses.
This base station allows for recharging the batteries or fuel cells in multiple dispensing devices and/or FOBs simultaneously. In addition to recharging the drug dispensing devices and FOBs, the base station may provide one or more of the following functionality: wireless or wired connectivity to a peripheral device, computer or network; feedback on the charging state for the devices being recharges; an interface for viewing, adding, deleting, or modifying the data on a drug dispensing device or FOB; a means for synchronizing data between multiple drug dispensing devices and/or FOBs; and a means for conducting a diagnostic test on drug dispensing devices and/or FOBs.

Single and Multiple Dose applicators
[00224] The invention provides disposable applicators for delivering dosage forms comprising sufentanil to the oral mucosa of a patient such that application to a pre-determined location for drug delivery (e.g. the mouth, sublingual space, etc.) is effected.
[0225] In one approach to the invention, a dosage form is delivered to the oral mucosa, using a single dose applicator. The dosage form is provided in a child-resistant drug dispensing device or packaging and delivered for example, to the sublingual cavity. The dosage form may be self-administered or alternatively, the dosage form is administered with assistance with or without a device.
[0226] In one embodiment, a single dose applicator (SDA) is used to a drug dosage forms, provided as a solid tablet, a liquid capsule, a gel capsule, a liquid, a gel, a powder, a film, a strip, a ribbon, a spray, a mist, a patch, or any other suitable drug dosage form.
[0227] The single dose applicator (SDA) may contain the dosage form within, may have the drug dosage form attached or affixed to it, may have the dosage form dissolved in it, and may afford a seal against moisture, humidity, and light.
The single dose applicator may be manually manipulated by a patient, healthcare provider, or other user to place the dosage form in the proper location for drug delivery.
[0228] In practicing the invention, a single- or multiple-dose applicator or drug dispensing device may be used to deliver tablets or other dosage forms into the hand, mouth, under the tongue, or to other locations appropriate for specific drug delivery needs.
[0229] In one embodiment, a single- or multiple-dose applicator or drug dispensing device is used to deliver a dosage form to the oral mucosa, e.g., the sublingual space.
[0230] The dosage forms inside the dispensing device remain dry prior to dispensing, at which point a single dosage form is dispensed from the device into the mouth, e.g., the sublingual space, wherein a patient's saliva will wet the tablet and allow for tablet disintegration/erosion and drug delivery.
[0231] The SDA may be provided as a pair of forceps, a syringe, a stick or rod, a straw, a pad, a capsule, a cup, a spoon, a strip, a tube, an applicator, a dropper, a patch, an adhesive pad, an adhesive film, a sprayer, an atomizer, or any other form suitable for the application of a single drug dosage form to the oral mucosa of a subject, e.g., the oral mucosa in the sublingual space. As will be understood by one of skill in the art, the SDA design may vary, so long as it is effective to place a drug dosage form, such as a tablet, in the desired location on an oral mucosal membrane, e.g., in the sublingual space, in a manner that preserves integrity of the drug dosage form in the dispensing process. After use, the SDA is disposed of, so as to eliminate the risk of contaminating the drug dispensing device with saliva, or other contaminants.
[0232] For sublingual administration, a small volume dosage form is administered by placement under the tongue, using an SDA, generally adjacent the frenulum.
[0233] The dosage form may be provided in a package that consists of molded plastic or laminate that has indentations (''blisters") into which a dosage form is placed, referred to herein as a "blister pack". A cover, typically a laminated material or foil, is used to seal to the molded part. A blister pack may or may not have pre-formed or molded parts and may be used to package an SDA of any type.
[0234] Such blister packs may be provided in a child resistant multiple drug dispenser (MDA), which may serve to dispense the dosage forms housed therein or may be used for storage of a plurality of SDAs.
[0235] Figs. 20A-C, Figs.21A-F, Figs. 23A-C and Figs. 24A and B are schematic depictions of exemplary embodiments of a SDA of the invention.
[0236] In one approach, the present invention provides disposable single dose applicators comprising a blister pack 151, which contains drug dosage forms 67 inside a housing and a handle 131, wherein a backing, such as a foil seal 135 covers the dosage form 67 and the handle 131, as shown for example in Fig. 21B
and 21D.
[0237] In one embodiment, the disposable single dose applicator, the combination of housing or tube 129 and handle 131 has the shape of a spoon.
[0238] The housing or tube 129 for the dosage form 67 is a blister pack 151 that accommodates a unit dose of a dosage form 67 for administration to a subject.
The dosage form 67 is sealed in the blister pack 151 by a foil or other type of seal 135.
[0239] In some embodiments, the foil or other type of seal 135 is removed prior to administration of the dosage form 67 and the handle 131 is used to place the dosage form 67 in the appropriate location against the oral mucosa of the subject such that the dosage form 67 adheres to the oral mucosa. See, e.g., Figs. 21B, 21D, 21E and 21F. In other embodiments, the foil or other type of seal 135 is perforated and removed prior to administration of the dosage form 67 by folding the applicator 123 at the perforation 149 prior to administration where the handle 131 is used to place the dosage form 67 in the appropriate location against the oral mucosa of a subject. See, e.g., Figs. 24A and B. This permits the handling of only a single drug dosage form 67 at a time and prevents the other individually sealed drug dosage forms 67 from becoming exposed to saliva, humidity and the like.
[0240] The foil or other type of seal 135 of a disposable applicator 123 including handle 131 is typically made of a single piece of foil laminate, paper, plastic or other covering, i.e. an applicator tab 147 that spans the back of the housing or tube 129 alone or both the housing or tube 129 and the handle 131, effectively seals the dosage form 67 in a blister pack 151 or other container.
[0241] The handle 131 enables proper placement of the dosage form 67 without touching the dosage form 67.
[0242] A plurality of single dose applicators may be provided as a series of individual single dose applicators attached by the backing or housed in multiple dose dispenser 137.
[0243] Figs. 14A and 14B show one embodiment of a single dose applicator 123 a dispensing device for delivering drug dosage forms. The dispensing device shown in Fig. 14A depicts the single dose applicator 123 that is ready to dispense a drug dosage form 67. In one aspect of this embodiment, a user pinches the single dose applicator 125 which opens the applicator and a drug dosage form 67 is dispensed as shown in Fig. 14B.
[0244] Figs. 15A - C show an embodiment of a single dose applicator 123 that is comprised of a applicator shaped as a tube 129, a stopper seal 127, a handle (e.g., an ergonomic handle), and a single dosage form 67. Fig 15A shows the single dose applicator 123 in its sealed configuration, prior to use. Fig 15B shows the single dose applicator 123 with its stopper seal 127 removed, forming an opening 133, and ready for use. Fig 15C shows the single dose applicator 123 tilted so as to dispense the dosage form 67 on the oral mucosa, e.g., in the sublingual space.
[0245] Figs. 21A - F show several alternate embodiments of the single dose applicator 123. In all of these figures the applicator seal 127 is broken and the applicator is tilted so as to drop the drug dosage form 67 adjacent an oral mucosal membrane in the mouth of a subject, e.g., under the tongue for sublingual dosage form placement. Fig. 21A shows a tube like applicator 129 with a handle 131 located axially under the tube 129. Fig. 21B shows an applicator formed as a thermoform or blister package 151 with a foil seal 135 that is peeled so as to open the applicator package 141 prior to placing the dosage form 67. Fig. 21C shows an applicator that is a tube 129 which is broken to break the seal prior to dosage form 67 placement. Fig. 21D shows a blister pack tube 151 type dosage form package 141 with a handle 131 such that after the seal 135 is peeled back the blister pack 151 can be held and tilted to place the drug dosage form 67, on an oral mucosal membrane. Figs. 21E and 21F show blister pack 151 type packaging with a handle 131 shaped like a flower or an animal, respectively, to be used for single dose applicator 123 designed for pediatric use. Other single dose applicator shapes could include cartoon characters, animals, super-heroes or other appropriate shapes for pediatric applications.
[0246] Figure 23A shows a flat rigid applicator 123 with a dosage form 67 adhered to one end, for example, by means of a rapidly dissolving ingestible adhesive material such that when the applicator end with the dosage form is placed under the tongue, the adhesive dissolves, the dosage form 67 is placed on an oral mucosal membrane, such as in the sublingual space, and the applicator can be removed. Fig 23B shows an applicator 123 made from a water permeable material, impregnated with drug, forming a material and dosage from matrix. When the impregnated end of this applicator 123 is placed under in the mouth on an oral mucosal membrane, the moisture in the saliva dissolves the drug and delivers it transmucosally. Figure 18C shows dissolving film dosage forms 145 and a dosage form package with a plurality of dissolving film dosage forms 143 within it.
The dissolving film dosage form 143 is removed from the package 141 and placed on an oral mucosal membrane, e.g., in the sublingual space where it dissolves and delivers the drug transmucosally.
[0247] Figs. 24A - B provides an illustrations of two stages of use of one embodiment of a single dose applicator 123. Figure 24A shows the applicator 123 in its configuration prior to use, with two applicator tabs 147, two perforations 149, and a blister pack 151 containing a dosage form 67. In order to administer the dosage form 67, the two applicator tabs 147 are bent downward at the perforations 149, forming a handle 131, and the seal 135 is peeled back to reveal the blister pack 151 and allow the dosage form 67 to be dropped on an oral mucosal membrane, e.g., in the sublingual space.
[0248] In another embodiment, a drug dispensing device of the invention may contain a plurality of SDA's, in a cartridge or individually packaged, and may dispense a single SDA containing a single drug dosage form for use by the patient, healthcare provider, or user. The drug dispensing device may dispense single SDA's in the same way and with the same features as would be advantageous for the dispensing of single drug dosage forms described in the invention.
[0249] In yet another embodiment the multiple dose applicator 137 is a device which comprises one or more drug dosage forms 67 or single dose applicators 123, a portable power means, like a battery, a printed circuit board, a data connectivity means, and a user interface. In this embodiment the drug dispensing device may include the ability to perform one or more of the following functions: record drug dosage dispensing history, check user identification by means of fingerprint identification, RFID, voice recognition, etc., allow the dosage history to be transferred to another device, computer or network, and/or provide a lockout period between dose dispenses.
[0250] Fig. 22 is a schematic depiction of an exemplary multiple dose applicator 137 for delivering dispensing drug dosage forms 67, each individually packaged in a single dose applicator 123.
[0251] Figs. 25A - D are schematic depictions of additional examples of single dose applicators (SDAs), including a tweezer or reverse scissor-type SDA
(25A), where drug dosage form 67 is held between the two sides 153 of the SDA 123 such that when the latch 19 is released, the drug dosage form 67 is no longer held by the SDA and can be placed on an oral mucosal membrane by the user; a syringe-type SDA (25B) with a circular channel, where a drug dosage form 67 is pushed out of the end of the channel when a user pushes 155, the slider or plunger 159; a pusher-type SDA (25C) with a rectangular channel where a drug dosage form 67 is pushed out of the end of the channel when a user pushes 155, the slider 159;
or a slider-type SDA (25D) where drug dosage form 67 is held in a pocket 161 and a drug dosage form 67 becomes accessible when a user pulls 157 a slider 159.
[0252] Figs. 26A - D provide a schematic depiction of a multiple dose applicator (MDA) 137 or container for storage of a plurality of SDAs 123 prior to use (26A);
where in the exemplified embodiment, there is a slot in the upper cover of the (MDA) 137 for removal of individual SDAs 123 (26B); such that each individual SDA

comprises a drug dosage form 67 (26C); and the SDA 123 facilitates placement of the drug dosage form 67 under the tongue in the sublingual space (26D).

IX. Methods and Systems for Delivering Small Volume Sufentanil Dosage Forms Using a Device.
[0253] Methods and systems for delivering small volume sufentanil-containing dosage forms using a device are provided. Fig 17 provides a schematic architecture connection diagram illustrating the various components that may be included in a dispensing device or system for dispensing small volume drug dosage forms, including a device with a separate head 13, body 15 and cartridge 17, a portable docking FOB 113, Patient RFID 115 and a base station 117.
[00254] A block diagram illustrating one aspect of communication in a drug dispensing system, including an RFID tag, a drug dispensing device, a base station/dock and a healthcare provider personal computer system wherein a drug dispensing device can communicate with the physician or care giver, via the dock, or by means of a wired or wireless communication method is provided in Fig. 18A.
[00255] A block diagram illustrating another aspect of communication in a drug dispensing system, including an RFID tag, a drug dispensing device, a portable docking FOB, a base station and a healthcare provider personal computer is provided in Fig. 18B. The drug dispensing device may communicate with the physician or care giver, via the FOB, by means of a wired or wireless communication method to provide usage information and information regarding the respiratory status or blood pressure of the patient to the physician at regular intervals. The FOB can be adapted to attach to a cord so as to allow the FOB to hang from the neck of the physician or caregiver.
[00256] Exemplary features of the dispensing device include the following:
In one embodiment, the head, body, and cartridge comprise the handheld portion of the device. This device assembly has a latch to disconnect the head and body, and a dispense button for patient use. The device also has lights to show lock-out status, errors, and power. In this embodiment, the cartridge which contains the drug dosage forms and the body are used a single time only.
[00257] The system may comprise a portable dock which is handheld, independent of the patient device and solely for healthcare professional use. The dock enables higher level feature use such as deeper queries into patient device use, the ability to upload device data, unlocking of the head/body and the tether, lockout override for dosing the patient, and a larger reading display. The dock is also used to setup and take down the patient device.
[00258] The system may also comprise an RFID bracelet that is activated via the dock and is worn by the patient to establish and control dosing to correct patient and to that patient alone. This feature prohibits use of the device by others.
[00259] The system may further comprise a recharging base used to charge the dock and heads and is also used to update the heads and docks when new software becomes available or when new users are programmed into the system.
[00260] The drug dosage forms are typically provided in disposable cartridges which are loaded into the device prior to administration.
[00261] Exemplary set-up instructions for the device include the following steps:
The device head and dock are charged on the recharging station.
The device body and wristband are removed from the packaging.
The device head and dock are removed from the charging station.
The cartridge is loaded into the body by inserting a cartridge into the device body as indicated ensuring that the cartridge "clicks' and is locked in place.
The device body (with cartridge) is assembled onto the head.
The power button on the assembled device is pushed to power-up the system.
The power button on the dock is pushed to power-up the dock.
The assembled device is plugged into the dock.
A healthcare professional scans their fingerprint or inputs a unique password in order to unlock the dock.
The device reads the label on the cartridge and the dock displays setup information, for example, the drug name, the quantity of tablets, the drug concentration, the preset lockout time, the duration of use (72 hours), and the battery status of the head.
After the information is read from the cartridge and displayed on the dock, the healthcare professional will be requested to confirm that all information is correct and will require a witness to verify the information.
The dock will require that the patient wristband be paired to the device by bringing the wristband close to the device.
The device will read the band and request confirmation of the band number;
selection and confirmation of the number The patient ID is entered into the dock. i.e. patient medical record number The wristband is placed on the patient's hand that will be used to operate device.
Then, the dock will indicate that it is ready to dispense a plastic initialization tablet or "shipping tablet".
Upon confirmation, the device will dispense a plastic initialization tablet or "shipping tablet". This step is used by the device to calibrate the dispensing mechanism, initiate the cartridge for use, and allows the healthcare professional to verify proper use and to train the patient with a "shipping" or placebo-type tablet.
Once the plastic initialization tablet or "shipping tablet" is dispensed, the dock will require the healthcare professional to confirm that the plastic tablet was dispensed.
After confirmation, the display will indicate that the device is ready for use.
[00262] In some cases, a tether can be connected to the device via the dock.
The dock will allow the healthcare professional to lock and unlock the tether as required.
[00263] If a patient will self administer a drug dosage form using the device, the patient will be trained prior to use.
[00264] Exemplary use of the claimed devices and systems is provided in Examples 6-8.
[00265] The following examples are provided to illustrate the invention and are not intended to limit any aspect of the invention as set forth above or in the claims below.
EXAMPLES
[00266] The following examples are provided to illustrate the invention and are not intended to limit any aspect of the invention as set forth above or in the claims below.
[00267] Two different sublingual sufentanil formulations were evaluated including a slower-eroding form (erosion time of approximately 15-25 minutes; Examples 1A
and 1B), and a faster-eroding form (approximate erosion time of 6-12 minutes;
Examples 2A and 2B). Patients were blocked with a mu-opioid receptor antagonist, naltrexone (50 mg orally twice per day).
[00268] Sufentanil plasma concentrations with respect to time were analyzed and tabulated. The maximum sufentanil concentration in plasma (Cmax), time to Cmax (Tmax) and terminal t1/Z were summarized for each dose group. After the final dosing of the repeat-dose studies, the sufentanil t1/Z was determined. Comparisons were made between the area under the curve (AUC) for each single administration of sublingual sufentanil dose vs. IV. Cmax, Tmax and t1/Z data were compared for each sublingual sufentanil dosage vs. IV and sublingual administration of sufentanil liquid.
Example 1: Evaluation of the Bioavailability and Pharmacokinetics following Sublingual Administration of a Small Volume Sufentanil Dosage Form.
[00269] Example 1A: All subjects received a 10-minute IV infusion of 5mcg sufentanil. After a 1-day washout period, each subject then received a single sublingual administration of a dosage form (comprising a slow-eroding formulation) containing 2.5mcg of sufentanil. On the two subsequent study days, the dose was escalated, and each subject received a dosage form (comprising a slow-eroding formulation) containing 5 and 10mcg of sufentanil.
[00270] Example 1B: All subjects received four repeated sublingual doses of a dosage form (comprising a slow-eroding formulation) containing 5mcg of sufentanil administered at 10-minute intervals.
[00271] The slow-eroding sublingual sufentanil formulation containing 10mcg sufentanil is provided below:

Ingredient Amount Sufentanil Citrate 0.27%
Mannitol (Pearlitol 200SD) 73.77%
PEG 8000 14.98%
Polyox 303 3.00%
Lutrol F68 2.00%
Stearic Acid 5.00%
Mg Stearate 1.00%
Total 100.00%
[00272] The sufentanil plasma concentration at various time points following a single sublingual administration of a 2.5, 5, or 10mcg sufentanil dosage form (slow-eroding) or 4 administrations of a 5mcg sufentanil dosage form (slow-eroding) minutes apart are shown in Figure 1.
[00273] The mean sufentanil ti/Z was similar for all of the sufentanil doses and varied from 1.56 hours (5mcg sublingual dosage form) to 1.97 hours (10mcg sublingual dosage form) with no obvious differences based on dose or route of administration (Table 1). The mean sufentanil Cmax and AUC;nf increased with dose and were proportional to dose. The Tmax following a single sublingual administration of sufentanil ranged from 0.68 to 0.77 hours. The bioavailability following sublingual administration varied from 74.5% in subjects who were administered 5mcg sufentanil dosage forms to 95.5% in subjects who were administered 10mcg sufentanil dosage forms.
[00274] Table 1 provides a summary of pharmacokinetic parameters including Cma, Tmaxi AUCinfi F and ti/Z. The Cmax after multiple sublingual dosing was 46.36 pg/mL.
The mean AUCinf increased with multiple sublingual dosing of sufentanil and was generally proportional to dose when compared to single sublingual administration.
The bioavailability of sufentanil following multiple sublingual dosing (97.2%) was greater than that following single administration at the same dose level (74.5%).
Table 1. Summary of Sufentanil Pharmacokinetic Parameters Parameter 5mcg IV 2.5mcg 5mcg lomcg 4x5mcg Cmax /mL 81.3 28.1 6.8 2.1 10.9 3.5 27.5 7.7 46.4 12.4 Tmax (hr) 0.16 0.03 0.73 0.13 0.77 0.29 0.68 0.22 1.16 0.23 AUCinf hr* /mL 38.4 8.5 18.0 4.5 27.4 9.1 71.2 20.7 146.5 39.1 tl Z (hr) 1.66 0.72 1.71 0.51 1.56 0.57 1.97 0.85 3.29 1.10 F % - 95.3 19.1* 74.5 26.3* 95.5 29.2* 97.2 21.2*
*%F calculated using 5mcg IV AUC
[00275] A paired t-test comparison of the mean sufentanil Cmax and AUCinf parameters was conducted after normalizing to the 10 mcg sublingual dose. The results are shown in Tables 2A and 2B. Results show that the Cmax and AUCinf were dose proportional from 2.5 to 10 mcg. Supporting data for dose-proportionality of the Cmax and AUCinf is shown in Figures 2 and 3, respectively.
Table 2A: Comparison of Sufentanil Pharmacokinetic Parameters Dose-Normalized to 10mc (2.5 mc slow-eroding dosage forms) Standard t p n = 12 2.5 mcg 10 mcg Difference deviation value value Cmax (pg/mL) 27.24 27.45 -0.21 10.24 -0.07 0.946 AUCinf hr* /mL 71.85 71.18 -0.67 16.31 0.14 0.89 Table 2B: Comparison of Sufentanil Pharmacokinetic Parameters Dose-Normalized to 10mcg 5mcg slow-eroding dosage forms) Standard t p n = 12 5 mcg 10 mcg Difference deviation value value Cmax (pg/mL) 21.81 27.45 -5.65 10.99 -1.78 0.10 AUCinf hr* /mL 54.85 71.18 -16.33 17.94 -3.15 0.009**
** p value <0.05, statistically significant
[00276] Mean sufentanil plasma concentrations versus time (+/- SD) following repeated sublingual administration of 4 x 5mcg sufentanil dosage forms (slow-eroding) at 10 minute intervals in healthy human volunteers as compared to IV
Infusion of 5mcg sufentanil over 10 minutes are shown in Fig. 4.
[00277] A simulation was used to estimate sufentanil plasma concentration following administration of 4 x 5mcg sublingual sufentanil dosage forms (slow-eroding), administered 10 minutes apart. The simulation was conducted by superposition of the mean plasma concentration over time profile of a single administration of the 5mcg sufentanil dosage form (slow-eroding). The simulation predicted and the observed mean ( SE) sufentanil plasma concentration versus time profiles were compared over a period of 12 hours (Fig. 5A) and a period of 2.5 hours (Fig. 5B). The predicted sufentanil concentrations based on the simulation closely tracks the observed sufentanil plasma concentration over time.

Example 2: Further Evaluation of the Bioavailability and Pharmacokinetics of Sufentanil following Sublingual Administration of a Small Volume Dosage Form.
[00278] Example 2A: Subjects were administered 5mcg of sufentanil solution via the sublingual route (N=2) or a 10-minute IV infusion of 5mcg sufentanil (N=10), a single sublingual administration of a dosage form containing 10mcg of sufentanil (faster-eroding formulation) and four repeated sublingual doses of a dosage form containing 10mcg of sufentanil (faster-eroding formulation) administered at 20-minute intervals.
[00279] Example 2B: All subjects were administered a 20-minute IV infusion of 50mcg sufentanil and a single sublingual administration of a dosage form containing 80mcg of sufentanil (faster-eroding formulation).
[00280] The fast-eroding sublingual sufentanil formulation containing 10mcg sufentanil is provided below:
Component Amount Sufentanil Citrate 0.26%
Mannitol SD100 70.64%
Di-Calcium Phosphate di-hydrate 20.00%
HPMC K4M Premium CR 3.00%
Stearic Acid 5.00%
Mg Stearate 1.00%
BHT 0.10%
Total 100.00%
[00281] The sufentanil plasma concentration (mean +/- SD) at various time points following a single sublingual administration of 10mcg and 80mcg sufentanil dosage forms (faster-eroding) and 4 administrations of the 10mcg sufentanil dosage form (faster-eroding) 20 minutes apart are shown in Figure 6.
[00282] The mean t1/2 was similar for the single sufentanil administrations and varied from 1.72 hours (5mcg IV) to 1.67 hours (10mcg sublingual). The mean sufentanil AUCinf increased with dose following single and multiple sublingual sufentanil administrations. The bioavailability was 60.9% in subjects treated with a single 10mcg sublingual sufentanil dosage form (fast-eroding) and 87.8%
following multiple (4 x 10mcg) sublingual sufentanil administrations.
[00283] A simulation was used to estimate sufentanil plasma concentration following administration of 4 x 10mcg sublingual sufentanil dosage forms (faster-eroding), administered 20 minutes apart. The simulation was conducted by superposition of the mean plasma concentration over time profile of a single administration of the 10mcg sufentanil dosage form (faster-eroding). The simulation-predicted and the observed mean ( SE) sufentanil plasma concentration versus time profiles were compared over a period of 12 hours (Fig. 7A) and a period of 2.5 hours (Fig. 7B). The observed sufentanil plasma concentrations were greater than the predicted sufentanil plasma concentrations (based on the simulation) over time.
Table 3. Summary of Sufentanil Pharmacokinetic Parameters Parameter 5mcg IV lOmc 4xlOmcg 80mc 50mc IV
Cmax (pg/mL) 63.9 28.2 16.5 6.8 78.7 20.1 127.2 42.3 561.1 277.7 Tmax (hr) 0.17 0.0 0.84 0.35 1.41 0.25 0.89 0.35 0.34 0.11 AUC;,,f hr* /mL 39.4 9.6 44.9 24.6 253.4 70.1 382.1 88.2 528.0 134.4 ti Z (hr) 1.72 0.47 1.67 0.67 3.54 1.02 4.23 0.90 3.69 0.78 F % - 60.9 27.7* 87.8 22.2* 70.1 20.1*
*%F calculated using 5mcg IV AUC
[00284] The bioavailability following sublingual administration of the 80mcg sufentanil dosage form (faster-eroding) was 70.1%.
[00285] A paired t-test comparison of the mean sufentanil Cmax and AUCinf parameters was conducted after normalizing to the 10 mcg sublingual dose. The results presented in Table 4 show that the Cmax and AUCinf were dose proportional from 10 to 80 mcg. Supporting data for dose-proportionality of the Cmax and AUCinf IS
shown in Figures 8 and 9, respectively.

Table 4: Comparison of Sufentanil Pharmacokinetic Parameters Dose-Normalized to 10mcg (80 mcg faster-eroding dosage forms) Standard t p n = 11 10 mcg 80 mcg Difference deviation value value CmaX (pg/mL) 16.59 16.93 -0.34 8.04 -0.14 0.89 AUC;,,f hr* /mL 45.02 50.88 -5.86 23.85 -0.81 0.43
[00286] A simulation of sufentanil concentrations following multiple administrations of 10 or 15mcg sublingual sufentanil dosage forms (slow-eroding), administered every 20 minutes was carried out. Dosage forms comprising the slow-eroding formulation resulted in a bioavailability of greater than 95%, and therefore serve as a basis for an estimate of the highest predicted steady-state sufentanil concentration following multiple administrations of 10 or 15mcg sublingual sufentanil dosage forms.
Steady-state sufentanil concentrations can be reached in about 12 hours after repeated sublingual doses at 20 minute intervals. The simulation predicted steady-state sufentanil concentrations of 200 pg/mL for administration of 10 mcg of sufentanil at 20 minute intervals and 300 pg/mL for administration of 15 mcg of sufentanil at 20 minute intervals as shown in Figures 10A and 10B, respectively. The simulations suggest that a minimal re-dosing interval of 20 minutes is safe.

Example 3. Acute Pain Management in the Outpatient Setting by Administering a Sufentanil-Containing Dosage Form Using a Device.
[00287] A pharmacist loads a drug dispensing device with a drug cartridge which includes 40 sufentanil dosage forms. Each cartridge has two colored initialization tablets (called "shipping tablets") arranged to be the first two tablets dispensed. The device has a means for loading the cartridge, which is either a port, hatch, or door that is secure and inaccessible to unauthorized users. Once the pharmacist has loaded the cartridge into the device, he locks the device access port, hatch or door.
The pharmacist then docks the dispensing device for the first time to a dock that is connected to a personal or other computer, using the docking connector, and then programs the device. Programming involves uploading the dosage strength of the dosage forms, the number of dosage forms loaded in the device, the prescribed frequency of dosage form usage, the number of dosage forms to be used per day, the current date and time, the preferred language, a valid thumbprint or other identification for identifying the patient, and the physician's identification information, in case the device is lost and found.
[00288] Once the dispensing device is programmed, the pharmacist demonstrates proper usage and tests the device by dispensing a single shipping tablet. The pharmacist then gives the dispensing device to the patient and observes the patient dispense a shipping tablet to ensure proper usage and functionality. Along with the dispensing device, the pharmacist provides the patient with a radio frequency identification (RFID) tag that must be within approximately 5 inches of the device to allow the dispensing device to operate.
[00289] When the patient wants to administer a dose of the drug, he or she will hold the dispensing device, and push any button to wake the device up from its sleep mode. The device will query the user for either a thumbprint reading or a personal identification number (PIN). The device will then search for a validated RFID key within range. Once these conditions are met, the dispensing device will query its internal memory and clock to make sure that the dosage regimen programmed by the pharmacist is not being violated by the current usage request.
At this point the device displays status information, such as the date and time, the number of doses left, the last time a dosage was used, the patient's name, etc., and the pharmacist informs the patient that the device is ready to dispense the dosage forms by a visual and/or audible signal.
[00290] The patient will hold the dispensing end of the device under his or her tongue and press the dispensing lever. When the dosage form is dispensed a tone will sound to inform the patient that the dosage form was properly delivered.
At this point the device will lock down to prevent further dispensing until the preprogrammed lock-out time has passed, at which time the device will be ready to use again.

Example 4: Acute Pain Management in the Inpatient Setting by administering a Sufentanil-Containing Dosage Form Using A Device.
[00291] A post operative patient requires acute pain treatment following surgery.
The surgeon prescribes oral transmucosal sufentanil to be administered using the drug dispensing device. The attending nurse takes the prescription order to the pharmacist or automated pharmaceutical inventory management system (e.g.
Pyxis) and obtains a sufentanil-containing drug cartridge for sublingual delivery.
The cartridge is labeled and equipped with an RFID electronic tag containing drug label information. The cartridge is labeled and equipped with an RFID electronic tag containing drug label information.
[00292] The nurse then takes a disposable dispensing portion of the drug dispensing device from inventory, and proceeds to a base station to obtain a reusable controller portion of the drug dispensing device that has completed its recharge cycle and is ready for use. The nurse inserts the drug cartridge into the disposable dispensing portion, and then affixes this to the reusable controller portion of the drug dispensing device and locks the disposable portion into the reusable portion of the drug dispensing device. At this point the device reads the RFID
tag on the drug cartridge and uploads the appropriate drug information, including the type of drug, the dosage strength, the programmed lockout period between doses, etc.
The nurse confirms the proper drug cartridge information has been read by the drug dispensing device and gives the drug dispensing device to the patient for patient controlled dispensing of the pain medication.
[00293] When the patient requires pain medication, she takes the drug dispensing device in her hand, and places the dispensing tip in her mouth, under her tongue and presses the dispense button. The drug dispensing device then does an internal check to ensure that the proper lockout period has elapsed since the last dosage dispense. At this point the drug dispensing device dispenses a dosage form under the patient's tongue and provides feedback that dosing was successful. The patient removes the drug dispensing device from her mouth and allows the sublingual dosage form to dissolve under her tongue. The patient may attempt to dispense as frequently as she desires, but the drug dispensing device will only allow successful dosing after the appropriate lockout period has elapsed. The drug dispensing device electronically logs the dispensing attempts and successful dispenses in its dosing history.
[00294] Periodically the nurse checks on the patient and drug dispensing device.
During such checks, the nurse inspects the drug dispensing device to see that there are no errors and to check the number of remaining dosage forms in the drug dispensing device, and returns it to the patient.
[00295] When the patient is discharged, the nurse takes the drug dispensing device and unlocks the reusable portion from the disposable portion, disposes of the cartridge and disposable portion of the drug dispensing device. The nurse then connects the reusable portion of the device to a computer and uploads the patient use information from the drug dispensing device to the computer for input into the patient's medical records. The nurse cleans the reusable controller portion and returns it to the base station for recharging.

Example 5: Acute Pain Management in the Inpatient Setting by administering a Sufentanil-Containing Dosage Form Using A Device and a Portable Dock.
[00296] A post operative patient requires acute pain treatment following surgery.
The surgeon prescribes oral transmucosal sufentanil to be administered using the drug dispensing device. The attending nurse takes the prescription order to the pharmacist or automated pharmaceutical inventory management system (e.g.
Pyxis) and obtains a sufentanil-containing drug cartridge for sublingual delivery.
The cartridge is labeled and equipped with an RFID electronic tag containing drug label information. The cartridge is labeled and equipped with an RFID electronic tag containing drug label information. The cartridge includes a shipping tablet or initialization tablet in the first to be dispensed location of the dosage form stack.
[00297] The nurse then takes a disposable dispensing portion of the drug dispensing device from inventory, and proceeds to a base station to obtain a reusable controller portion of the drug dispensing device that has completed its recharge cycle and is ready for use. The nurse inserts the drug cartridge into the disposable dispensing portion, and then affixes this to the reusable controller portion of the drug dispensing device. Next, the nurse takes a portable dock (or docking FOB) from the base station where it has been recharging, and docks the assembled drug dispensing device to the portable dock. The portable dock and the assembled drug dispensing device communicate electronically and a setup menu comes up on the portable dock for setting up the drug dispensing device.
[00298] At this point the device locks the reusable and disposable portions together, reads the RFID tag on the drug cartridge and uploads the appropriate drug information, including the type of drug, the dosage strength, the lockout period between doses, etc. The dispensing device writes a code to the RFID tag on the cartridge identifying it as a used cartridge. The nurse enters her fingerprint in the fingerprint reader on the portable dock to gain secured access and proceeds to set up the drug dispensing device for use. The set up procedure includes entering user identification, the nurse's identification, confirming the proper time on the device, and confirming the proper drug cartridge information. The nurse then takes a disposable RFID bracelet and places this adjacent to the drug dispensing device at which point the drug dispensing device reads the tag and the nurse confirms that the proper bracelet tag has been read.
[00299] The nurse then confirms proper setup of the drug dispensing device by pressing the dispensing button once. The drug dispensing device actuates, dispensing the shipping tablet facsimile into the nurses hand, confirming proper operation. The drug dispensing device detects the dispensing of the shipping tablet, allowing for an internal system check of proper operation and internal calibration of the newly assembled system. If the internal dispensing check is successful, the portable dock queries the nurse to confirm that the shipping table was properly dispensed, and the nurse confirms the proper setup. The nurse then disengages the drug dispensing device from the portable dock, and proceeds to the patient's bedside for the final steps of setup.
[00300] The nurse places the RFID bracelet on the patient's wrist and affixes a theft resistant tether to the patient's bed and the other end to the drug dispensing device. The nurse then instructs the patient on proper use of the sublingual drug dispensing device, and gives the drug dispensing device to the patient for patient controlled dispensing of sufentanil.
[00301] When the patient requires pain medication, she takes the drug dispensing device in her hand, and places the dispensing tip in her mouth, under her tongue and presses the dispensing button. The drug dispensing device then does an internal check to ensure that the proper lockout period has elapsed since the last dosage dispense, and that the patient's RFID bracelet is present and readable. At this point the drug dispensing device dispenses a dosage form under the patient's tongue and provides a feedback that dosing was successful. The patient removes the drug dispensing device from her mouth and allows the sublingual dosage form to dissolve under her tongue. The patient may attempt to dispense as frequently as she desires, but the drug dispensing device will only allow successful dosing after the appropriate lockout period has elapsed. The drug dispensing device electronically logs the dispensing attempts and successful dispenses in its dosing history.
[00302] Periodically the nurse checks on the patient and device. During such a patient check in the nurse brings a portable docking FOB and docks the device to the FOB. The electronic connection enables the nurse to download the information from the drug dispensing device to the FOB. This information includes the use history, drug information, number of remaining dosage forms and duration of use since initial set up. The nurse then enters her fingerprint in the finger print scanner to gain access to the information and to drug dispensing device. Because the patient is requiring an additional dose of drug prior to the lockout period expiring, the nurse overrides the lockout period and then returns the drug dispensing device to the patient at which point the patient is able to take another dose.
[00303] The nurse leaves the patient's room with the portable docking FOB and returns to the nurse's station to record the dosing history in the patient's records.
When finished the nurse returns the FOB to the base station for recharging.
[00304] When the patient has used all of the dosage forms in the drug dispensing device, the nurse brings the portable docking FOB into the patient's room and docks the drug dispensing device to the FOB. The nurse then enters her fingerprint in the fingerprint scanner on the FOB to gain secured access to the drug dispensing device.
Next, the nurse unlocks the security tether and disconnects the drug dispensing device from the bed. She then unlocks the drug dispensing device and removes it from the FOB for disassembly. The nurse disconnects the disposable portion from the reusable portion, and removes the cartridge from the disposable portion. The nurse disposes of the disposable portion and the cartridge, and wipes the reusable controller portion with an antiseptic wipe to clean it before returning it to the base station. The reusable controller portion requires that the nurse return it to the base station where it recharges and runs an internal diagnostic test before being ready for use again.
[00305] The nurse then proceeds to set up a new drug dispensing device as described above and provides this to the patient.
[00306] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity and understanding, it will be apparent to those skilled in the art that certain changes and modifications may be practiced. Various aspects of the invention have been achieved by a series of experiments, some of which are described by way of the following non-limiting examples. Therefore, the description and examples should not be construed as limiting the scope of the invention, which is delineated by the appended description of exemplary embodiments.

Claims (20)

1. A dosage form for oral transmucosal administration to a subject, comprising:
from about 5 to about 100 micrograms (mcg) of sufentanil and a bioadhesive material, wherein said bioadhesive material provides for adherence to the oral mucosa of said subject and said dosage form has a volume of less than 30 microliters or a mass of less than 30mg.
2. The drug dosage form according to Claim 1, wherein said dosage form comprises a dose of sufentanil selected from the group consisting of 5mcg, 10mcg, 15mcg, 20mcg, 30mcg, 40mcg, 50mcg, 60mcg, 70mcg, 80mcg and 100mcg.
3. The dosage form according to Claim 1 or 2, wherein said dosage form has a mass of less than 10mg or a volume of less than 10µl.
4. The dosage form according to any one of Claims 1 to 3, wherein said oral transmucosal administration is sublingual or buccal administration.
5. The dosage form according any one of Claims 1 to 4, wherein the erosion time of said dosage form is from 30 seconds up to a time selected from the group consisting of minutes, 10 minutes, 15 minutes and 30 minutes.
6. The drug dosage form according to Claim 4 or 5, wherein said dosage form delivers at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or at least 99% of the total amount of sufentanil in said dosage form via the sublingual route.
7. The drug dosage form according to Claim 4, 5 or 6, wherein a single sublingual administration of said dosage form to a subject results in a bioavailability of greater than 50%.
8. The dosage form according to Claim 4, 5 or 6, wherein a single sublingual administration of said dosage form to a subject results in a bioavailability of greater than 60%.
9. The dosage form according to Claim 4, 5 or 6, wherein a single sublingual administration of said dosage form to a subject results in a bioavailability of greater than 70%.
10. The drug dosage form according to any one of Claims 4 to 9, wherein a single sublingual administration of said dosage form to a subject results in a T max with a coefficient of variation of less than 40%.
11. The drug dosage form according to any one of Claims 4 to 10, wherein following repeated sublingual administrations of said dosage form to a subject, the bioavailability is greater than the bioavailability following a single sublingual administration to said subject.
12. The drug dosage form according to any one of Claims 4 to 11, wherein the difference between the T max following repeated sublingual administrations and the time of the previous sublingual administration is shorter than the T max following a single sublingual administration to said subject.
13. The drug dosage form according to any one of Claims 4 to 12, wherein repeated sublingual administration of said dosage form to a subject results in a T max with a coefficient of variation of less than 40%.
14. The formulation according to any one of Claims 1 to 13, wherein said dosage form is selected from the group consisting of a lozenge, a pill, a tablet, a membrane and a strip.
15. The dosage form according to Claim 14, wherein said dosage form is a tablet.
16. A single dose applicator (SDA), comprising a dosage form according to any one of Claims 1 to 15.
17. The use of a single dose applicator (SDA) according to any one of Claims 1 to 16, for the treatment of pain.
18. The use according to Claim 17, wherein said pain is acute pain, breakthrough pain or post-operative pain.
19. The use according to Claim 18, wherein said pain is acute inpatient pain.
20. The use according to Claim 18, wherein said pain is post-operative pain.
CA2673856A 2007-01-05 2007-12-28 Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain Active CA2673856C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US11/650,174 2007-01-05
US11/650,174 US8202535B2 (en) 2006-01-06 2007-01-05 Small-volume oral transmucosal dosage forms
US11/985,162 2007-11-14
US11/985,162 US8865743B2 (en) 2006-01-06 2007-11-14 Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain
PCT/US2007/089018 WO2008085765A2 (en) 2007-01-05 2007-12-28 Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain

Publications (2)

Publication Number Publication Date
CA2673856A1 true CA2673856A1 (en) 2008-07-17
CA2673856C CA2673856C (en) 2018-11-27

Family

ID=39609243

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2673856A Active CA2673856C (en) 2007-01-05 2007-12-28 Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain

Country Status (10)

Country Link
US (5) US8865743B2 (en)
EP (1) EP2114383B1 (en)
JP (1) JP2010532745A (en)
KR (1) KR101545754B1 (en)
AT (1) ATE474564T1 (en)
CA (1) CA2673856C (en)
DE (1) DE602007007991D1 (en)
DK (1) DK2114383T3 (en)
PT (1) PT2114383E (en)
WO (1) WO2008085765A2 (en)

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8357114B2 (en) 2006-01-06 2013-01-22 Acelrx Pharmaceuticals, Inc. Drug dispensing device with flexible push rod
US8535714B2 (en) 2006-01-06 2013-09-17 Acelrx Pharmaceuticals, Inc. Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain
US9066847B2 (en) 2007-01-05 2015-06-30 Aceirx Pharmaceuticals, Inc. Storage and dispensing devices for administration of oral transmucosal dosage forms
US8753308B2 (en) 2006-01-06 2014-06-17 Acelrx Pharmaceuticals, Inc. Methods for administering small volume oral transmucosal dosage forms using a dispensing device
US8202535B2 (en) 2006-01-06 2012-06-19 Acelrx Pharmaceuticals, Inc. Small-volume oral transmucosal dosage forms
US8865743B2 (en) 2006-01-06 2014-10-21 Acelrx Pharmaceuticals, Inc. Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain
US9289583B2 (en) 2006-01-06 2016-03-22 Acelrx Pharmaceuticals, Inc. Methods for administering small volume oral transmucosal dosage forms using a dispensing device
US8252328B2 (en) 2006-01-06 2012-08-28 Acelrx Pharmaceuticals, Inc. Bioadhesive drug formulations for oral transmucosal delivery
US8252329B2 (en) 2007-01-05 2012-08-28 Acelrx Pharmaceuticals, Inc. Bioadhesive drug formulations for oral transmucosal delivery
AU2008216867B2 (en) 2007-02-09 2014-01-30 Durect Corporation Transoral dosage forms comprising sufentanil and naloxone
JP5632284B2 (en) * 2007-08-07 2014-11-26 エーセルアールエックス ファーマシューティカルズ, インコーポレイテッド Compositions and methods for sedation and analgesia during treatment using oral transmucosal dosage forms
US8945592B2 (en) * 2008-11-21 2015-02-03 Acelrx Pharmaceuticals, Inc. Sufentanil solid dosage forms comprising oxygen scavengers and methods of using the same
WO2010107761A1 (en) * 2009-03-18 2010-09-23 Acelrx Pharmaceuticals, Inc. Improved storage and dispensing devices for administration of oral transmucosal dosage forms
US20110050412A1 (en) * 2009-08-18 2011-03-03 Cynthia Wittman Voice activated finding device
US20110091544A1 (en) * 2009-10-16 2011-04-21 Acelrx Pharmaceuticals, Inc. Compositions and Methods for Mild Sedation, Anxiolysis and Analgesia in the Procedural Setting
US20110218595A1 (en) * 2010-03-04 2011-09-08 Mcmillan William C Prescription device controller
US20110288128A1 (en) 2010-05-21 2011-11-24 Acelrx Pharmaceuticals, Inc. Oral Transmucosal Administration of Sufentanil
GB201014783D0 (en) * 2010-09-06 2010-10-20 St George S Hospital Medical School Apparatus and method for positioning a probe for observing microcirculation vessels
US20130020351A1 (en) * 2011-07-21 2013-01-24 Gojo Industries, Inc. Dispenser with optical keying system
NZ701428A (en) 2012-05-02 2016-10-28 Orexo Ab New alfentanil composition for the treatment of acute pain
CN105007904A (en) * 2014-02-07 2015-10-28 科学实验室药物公司 All natural, non-toxic sublingual drug delivery systems
TWI689326B (en) * 2014-08-06 2020-04-01 加拿大商複製細胞生命科學公司 Injection devices
SG11201704797TA (en) 2014-12-23 2017-07-28 Acelrx Pharmaceuticals Inc Systems, devices and methods for dispensing oral transmucosal dosage forms
FR3032353B1 (en) 2015-02-06 2017-03-10 Jacques Seguin PHARMACEUTICAL COMPOSITION AND DEVICE FOR THE TREATMENT OF PAIN
SE539111C2 (en) * 2015-06-03 2017-04-11 Iconovo Ab Single dose dry powder inhaler
ES2947769T3 (en) 2015-10-30 2023-08-18 Vertical Pharmaceuticals Llc Apparatus and methods for dispensing oral transmucosal dosage forms
JP2017112241A (en) * 2015-12-17 2017-06-22 ルネサスエレクトロニクス株式会社 Semiconductor device
US9731103B1 (en) 2017-01-13 2017-08-15 Berkshire Biomedical, LLC Computerized oral prescription administration devices and associated systems and methods
US10792226B2 (en) 2017-06-07 2020-10-06 Berkshire Biomedical, LLC Refill and dosage management devices and associated systems and methods for use with computerized oral prescription administration devices
US10441509B2 (en) 2018-03-16 2019-10-15 Berkshire Biomedical, LLC Computerized oral prescription administration with refillable medication dispensing devices and associated systems and methods
US10597206B2 (en) 2018-06-15 2020-03-24 Kenneth Corey Medicine container cover
US10729860B1 (en) 2019-05-22 2020-08-04 Berkshire Biomedical, LLC Computerized oral prescription administration for securely dispensing a medication and associated systems and methods

Family Cites Families (263)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2621655A (en) 1951-10-11 1952-12-16 Kenneth L Gothard Tablet dispensing gun
US3162322A (en) 1962-03-29 1964-12-22 John E Gilbertson Sanitary dispenser for cartridges holding fluid medicants
US3238941A (en) 1963-06-19 1966-03-08 Frost Eng Dev Balling gun
GB1142325A (en) 1965-05-14 1969-02-05 Higham Stanley Russell Means for administering drugs
US3757781A (en) 1971-09-17 1973-09-11 R Smart Tool for administering pills to animals
US3780735A (en) 1972-02-03 1973-12-25 J Crouter Dose syringe
US3789845A (en) 1973-01-29 1974-02-05 J Long Capsule and tablet administration for animals
FR2278317A1 (en) 1974-07-19 1976-02-13 Commissariat Energie Atomique ORAL IMPLANT FOR ADMINISTERING SOLUBILIZABLE PRODUCTS
US4060083A (en) 1976-04-01 1977-11-29 Hanson Raymond L Pill gun
US4237884A (en) 1978-03-17 1980-12-09 Victor Erickson Medication dispenser
GB2042888B (en) 1979-03-05 1983-09-28 Teijin Ltd Preparation for administration to the mucosa of the oral or nasal cavity
US4229447A (en) 1979-06-04 1980-10-21 American Home Products Corporation Intraoral methods of using benzodiazepines
US4465191A (en) 1982-06-14 1984-08-14 Darbo Rolf E Personal tablet dispenser
US4474308A (en) 1982-09-30 1984-10-02 Michel Bergeron Tablet ejector
US4489853A (en) 1982-12-15 1984-12-25 Korte Jay S Pill dispensing device
GB8332556D0 (en) 1983-12-06 1984-01-11 Reckitt & Colmann Prod Ltd Analgesic compositions
US5855908A (en) 1984-05-01 1999-01-05 University Of Utah Research Foundation Non-dissolvable drug-containing dosage-forms for use in the transmucosal delivery of a drug to a patient
US4863737A (en) 1985-05-01 1989-09-05 University Of Utah Compositions and methods of manufacture of compressed powder medicaments
US5288498A (en) 1985-05-01 1994-02-22 University Of Utah Research Foundation Compositions of oral nondissolvable matrixes for transmucosal administration of medicaments
DE3421468A1 (en) 1984-06-08 1985-12-19 Dr. Rentschler Arzneimittel Gmbh & Co, 7958 Laupheim LIPID NANOPELLETS AS A CARRIER SYSTEM FOR MEDICINAL PRODUCTS FOR PERORAL USE
EP0166315B1 (en) 1984-06-19 1989-08-23 BASF Aktiengesellschaft Gastro-resistant cylindrical pancreatine-microtablets
US5132114A (en) 1985-05-01 1992-07-21 University Of Utah Research Foundation Compositions and methods of manufacture of compressed powder medicaments
US4671953A (en) * 1985-05-01 1987-06-09 University Of Utah Research Foundation Methods and compositions for noninvasive administration of sedatives, analgesics, and anesthetics
US5122127A (en) 1985-05-01 1992-06-16 University Of Utah Apparatus and methods for use in administering medicaments by direct medicament contact to mucosal tissues
US5288497A (en) 1985-05-01 1994-02-22 The University Of Utah Compositions of oral dissolvable medicaments
US5785989A (en) 1985-05-01 1998-07-28 University Utah Research Foundation Compositions and methods of manufacturing of oral dissolvable medicaments
US4764378A (en) 1986-02-10 1988-08-16 Zetachron, Inc. Buccal drug dosage form
DE3632546A1 (en) 1986-09-25 1988-03-31 Knoll Ag DOSAGE DISPENSER
US4785969A (en) 1986-11-10 1988-11-22 Pyxis Corporation Medication dispensing system
DE3855054T2 (en) 1987-05-26 1996-07-18 Sumitomo Pharma Device for the administration of solid preparations
US4915948A (en) 1987-08-31 1990-04-10 Warner-Lambert Company Tablets having improved bioadhesion to mucous membranes
FR2619718B1 (en) 1987-09-02 1991-07-12 Medibrevex NOVEL GALENIC FORMS OF BETA-2-MIMETICS FOR PER- AND SUBLINGUAL ADMINISTRATION
US4873076A (en) 1988-04-29 1989-10-10 Baker Cummins Pharmaceuticals, Inc. Method of safely providing anesthesia or conscious sedation
DE68908527T2 (en) 1988-04-29 1993-12-16 Baker Norton Pharma METHOD FOR SAFE ANESTHESIA OR CONSCIOUSNESS EDITING.
JPH02121039A (en) 1988-10-31 1990-05-08 Toshiba Corp Load prediction simulation system for computer system
US5236714A (en) 1988-11-01 1993-08-17 Alza Corporation Abusable substance dosage form having reduced abuse potential
US5112616A (en) 1988-11-30 1992-05-12 Schering Corporation Fast dissolving buccal tablet
AT391300B (en) 1989-04-12 1990-09-10 Hinterreiter Ignaz TABLET DISPENSER
US5178878A (en) 1989-10-02 1993-01-12 Cima Labs, Inc. Effervescent dosage form with microparticles
US5223264A (en) 1989-10-02 1993-06-29 Cima Labs, Inc. Pediatric effervescent dosage form
US5190185A (en) 1990-05-18 1993-03-02 Baxter International Inc. Medication transport and dispensing magazine
GB9015522D0 (en) 1990-07-13 1990-08-29 Braithwaite Philip W Inhaler
EP0576605A4 (en) 1991-03-19 1994-06-08 Vithal J Rajadhyaksha Compositions and method comprising aminoalcohol derivatives as membrane penetration enhancers
US5296234A (en) 1991-10-11 1994-03-22 Abbott Laboratories Holder and packaging for a hardened medicated matrix
US5263596A (en) 1991-12-02 1993-11-23 Williams David R Medication dispenser station sub-assembly
US5348158A (en) 1991-12-19 1994-09-20 G. D. Searle & Co. Dispenser pack for the successive dispensing of a drug
NL9200844A (en) 1992-05-13 1993-12-01 De Wijdeven Gijsbertus G P Van DEVICE AND METHOD FOR INJECTING WITH A SOLID SUBSTANCE.
US5352680A (en) * 1992-07-15 1994-10-04 Regents Of The University Of Minnesota Delta opioid receptor antagonists to block opioid agonist tolerance and dependence
US5292307A (en) 1992-09-16 1994-03-08 Dolzine Theodore W Dispensing package for unit dosage
US5724957A (en) 1993-01-29 1998-03-10 Aradigm Corporation Intrapulmonary delivery of narcotics
US5507277A (en) 1993-01-29 1996-04-16 Aradigm Corporation Lockout device for controlled release of drug from patient-activateddispenser
US5694919A (en) 1993-01-29 1997-12-09 Aradigm Corporation Lockout device for controlled release of drug from patient-activated dispenser
US5489689A (en) 1993-09-30 1996-02-06 Mallinckrodt Chemical, Inc. Preparation of piperidine derivatives
US5366113A (en) 1993-12-13 1994-11-22 Kim David S Pill dispenser
US5489025A (en) 1994-03-01 1996-02-06 Romick; Jerome M. Unit-dose medication dispenser and multiple-dispenser frame therefor
US5507807A (en) 1994-03-01 1996-04-16 Shippert; Ronald D. Apparatus for the release of a substance within a patient
US5488953A (en) * 1994-04-15 1996-02-06 Ecocath, Inc. Diffracting doppler-transducer
US6328159B1 (en) 1994-07-13 2001-12-11 Centrix, Inc Single patient dose medicament dispenser with applicator
US6685013B2 (en) 1994-07-13 2004-02-03 Centrix, Inc. Single patient dose medicament dispenser with applicator
US5660273A (en) 1994-07-13 1997-08-26 Centrix, Inc. Single patient dose medicament dispenser with applicator
HU219117B (en) 1994-09-16 2001-02-28 Laboratoire Glaxo Wellcome S.A. Inhalation device
US5584805A (en) 1995-03-17 1996-12-17 Sutton; Matthew R. Animal pill-dispenser gun
US6881208B1 (en) 1995-06-05 2005-04-19 Joseph B. Phipps Method and device for transdermal electrotransport delivery of fentanyl and sufentanil
US6216033B1 (en) 1996-05-22 2001-04-10 Alza Corporation Device for transdermal electrotransport delivery of fentanyl and sufentanil
PT836511E (en) 1995-06-05 2004-08-31 Alza Corp DEVICE FOR DISTRIBUTION OF FENTANIL AND SUFENTANIL BY PERCUTANEOUS ELECTROTRANSPORTS
US6425892B2 (en) 1995-06-05 2002-07-30 Alza Corporation Device for transdermal electrotransport delivery of fentanyl and sufentanil
JPH11500646A (en) 1995-06-07 1999-01-19 オニール,アレクサンダー・ジョージ・ブライアン Patient controlled drug delivery device
US5984888A (en) 1995-06-07 1999-11-16 Kimberly-Clark Worldwide, Inc. Applicator and coating
US5710551A (en) 1995-07-26 1998-01-20 Ridgeway; Donald G. Self-medication monitoring system
CA2184316A1 (en) 1995-09-12 1997-03-13 Wei-Chi Liao Buccal delivery system for therapeutic agents
AUPN603895A0 (en) 1995-10-19 1995-11-09 University Of Queensland, The Production of analgesic synergy by co-administration of sub-analgesic doses of two strong opioids
GB2309966A (en) 1996-02-07 1997-08-13 Harwill Ind Pty Ltd Teat device for administering medicaments
US5995938A (en) 1996-02-27 1999-11-30 Whaley; Susan S. Medication compliance system
DE69722203T2 (en) 1996-07-05 2004-04-01 Novo Nordisk A/S DEVICE FOR INSERTING A PIG
NL1003596C2 (en) 1996-07-15 1997-05-27 Cons Health Entrepreneurs Bv Vending machine dispenser.
US6248789B1 (en) 1996-08-29 2001-06-19 Stuart L. Weg Administration of ketamine to manage pain and to reduce drug dependency
US5800832A (en) 1996-10-18 1998-09-01 Virotex Corporation Bioerodable film for delivery of pharmaceutical compounds to mucosal surfaces
US5954641A (en) 1997-09-08 1999-09-21 Informedix, Inc. Method, apparatus and operating system for managing the administration of medication and medical treatment regimens
US5752620A (en) 1996-11-13 1998-05-19 Pearson Ventures, L.L.C. Medication dispenser
US6529446B1 (en) * 1996-12-20 2003-03-04 Telaric L.L.C. Interactive medication container
US5968547A (en) 1997-02-24 1999-10-19 Euro-Celtique, S.A. Method of providing sustained analgesia with buprenorphine
US5950632A (en) 1997-03-03 1999-09-14 Motorola, Inc. Medical communication apparatus, system, and method
US6024981A (en) 1997-04-16 2000-02-15 Cima Labs Inc. Rapidly dissolving robust dosage form
US5945651A (en) 1997-07-17 1999-08-31 Chorosinski; Leonard Remotely programmable medication dispensing system
JPH1147233A (en) 1997-07-30 1999-02-23 Teikoku Seiyaku Co Ltd Plaster agent and preservation method thereof
US5850937A (en) 1997-08-14 1998-12-22 Rauche; Stephen J. Dispenser with means for alerting a user
EP1017370B1 (en) 1997-09-11 2003-10-29 Nycomed Danmark ApS MODIFIED RELEASE MULTIPLE-UNITS COMPOSITIONS OF NON-STEROID ANTI-INFLAMMATORY DRUG SUBSTANCES (NSAIDs)
US6660295B2 (en) 1997-09-30 2003-12-09 Alza Corporation Transdermal drug delivery device package with improved drug stability
US20030185761A1 (en) 1997-10-01 2003-10-02 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating pain
IL134395A (en) 1997-10-03 2005-08-31 Warner Lambert Co Compressed nitrogylycerin tablet and its method of manufacture
WO2000066458A1 (en) 1997-10-30 2000-11-09 HALLIN, Cristian under the firm of TELUM PRODUCTI N A tablet dispenser
US5997518A (en) 1998-01-14 1999-12-07 Laibovitz; Robert A. Apparatus and method for delivery of small volumes of liquid
KR20010040398A (en) * 1998-01-23 2001-05-15 마가렛 에이.혼 Chemically modified mutant enzymes and methods for producing them and screening them
US5981552A (en) 1998-10-23 1999-11-09 Taylor Pharmaceuticals Sublingual and buccal compositions of droperidol and method for treating migraine
US6350470B1 (en) 1998-04-29 2002-02-26 Cima Labs Inc. Effervescent drug delivery system for oral administration
US6200604B1 (en) 1998-03-27 2001-03-13 Cima Labs Inc. Sublingual buccal effervescent
US20030091629A1 (en) 1998-03-27 2003-05-15 Cima Labs Inc. Sublingual buccal effervescent
US6576250B1 (en) 1998-03-27 2003-06-10 Cima Labs Inc. Pharmaceutical compositions for rectal and vaginal administration
US6974590B2 (en) 1998-03-27 2005-12-13 Cima Labs Inc. Sublingual buccal effervescent
US6039251A (en) 1998-04-16 2000-03-21 Holowko; Paul L. Method and system for secure control of a medical device
DE69928267T2 (en) 1998-08-28 2006-06-08 Glaxo Group Ltd., Greenford DONOR
SE9803240D0 (en) 1998-09-24 1998-09-24 Diabact Ab A pharmaceutical composition having a rapid action
SE9803239D0 (en) 1998-09-24 1998-09-24 Diabact Ab Composition for the treatment of acute pain
JP2000142841A (en) 1998-11-17 2000-05-23 Lion Corp Container for small-sized article
JP3459882B2 (en) 1998-11-30 2003-10-27 株式会社バンダイ Article removal device
GB9826192D0 (en) 1998-12-01 1999-01-20 Controlled Theraputics Scotlan Oral transmucosal delivery
US6131765A (en) 1999-01-08 2000-10-17 Barry; William Dean Device for storing and dispensing solid-form medication
AU2879100A (en) 1999-02-12 2000-08-29 Miles A. Libbey Iii Formulation and system for intra-oral delivery of pharmaceutical agents
US6680071B1 (en) 1999-03-03 2004-01-20 R. P. Scherer Technologies, Inc. Opioid agonist in a fast dispersing dosage form
US6541021B1 (en) 1999-03-18 2003-04-01 Durect Corporation Devices and methods for pain management
US6835194B2 (en) 1999-03-18 2004-12-28 Durect Corporation Implantable devices and methods for treatment of pain by delivery of fentanyl and fentanyl congeners
US6234343B1 (en) 1999-03-26 2001-05-22 Papp Enterprises, Llc Automated portable medication radial dispensing apparatus and method
US6601729B1 (en) 1999-03-26 2003-08-05 Papp Enterprises, Llc Automated portable medication radial dispensing apparatus and method using a carrier tape
US6210699B1 (en) 1999-04-01 2001-04-03 Watson Pharmaceuticals, Inc. Oral transmucosal delivery of drugs or any other ingredients via the inner buccal cavity
US6762684B1 (en) 1999-04-19 2004-07-13 Accutrak Systems, Inc. Monitoring system
US6319510B1 (en) 1999-04-20 2001-11-20 Alayne Yates Gum pad for delivery of medication to mucosal tissues
US6190326B1 (en) 1999-04-23 2001-02-20 Medtrac Technologies, Inc. Method and apparatus for obtaining patient respiratory data
DE60044196D1 (en) 1999-04-29 2010-05-27 Peter James Brian Lamb APPARATUS FOR TRANSFERRING A NON-FLOWING AGENT IN A VAGINA
GB9911388D0 (en) 1999-05-18 1999-07-14 Glaxo Group Ltd Dispenser
US6395300B1 (en) 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US7919119B2 (en) 1999-05-27 2011-04-05 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
WO2001012174A1 (en) 1999-08-13 2001-02-22 Vela Pharmaceuticals Inc. Cyclobenzaprine for treating generalized anxiety disorder and compositions thereof
US6264983B1 (en) 1999-09-16 2001-07-24 Rhodia, Inc. Directly compressible, ultra fine acetaminophen compositions and process for producing same
FR2799124B1 (en) 1999-10-01 2004-08-13 Sanofi Synthelabo USE OF ANTAGONISTS OF CENTRAL CANNABINOID RECEPTORS FOR THE PREPARATION OF DRUGS
US6382204B1 (en) 1999-10-14 2002-05-07 Becton Dickinson And Company Drug delivery system including holder and drug container
US6572891B1 (en) 1999-10-23 2003-06-03 Alkaloid Ad Sublingual oral dosage form
US6264981B1 (en) * 1999-10-27 2001-07-24 Anesta Corporation Oral transmucosal drug dosage using solid solution
US7074935B2 (en) 1999-12-06 2006-07-11 Mallinckrodt Inc. Methods for the syntheses of alfentanil, sufentanil and remifentanil
CO5270018A1 (en) 1999-12-11 2003-04-30 Glaxo Group Ltd MEDICINAL DISTRIBUTOR
AT408488B (en) 1999-12-22 2001-12-27 Scan Messtechnik Gmbh MINIATURIZED SPECTROMETER
US6294999B1 (en) 1999-12-29 2001-09-25 Becton, Dickinson And Company Systems and methods for monitoring patient compliance with medication regimens
US6471645B1 (en) 1999-12-30 2002-10-29 Medtronic, Inc. Communications system for an implantable device and a drug dispenser
AU2001234005B2 (en) 2000-02-28 2006-01-19 Pharmakodex Limited Improvements in or relating to the delivery of oral drugs
US6364158B1 (en) 2000-03-24 2002-04-02 John Dimoulis Anesthetic dispensing station and method of using same
AU2001262992A1 (en) 2000-05-10 2002-02-18 University Of Kentucky Research Foundation System and method for intranasal administration of opioids
DE10025946A1 (en) 2000-05-26 2001-11-29 Gruenenthal Gmbh drug combination
AT409366B (en) 2000-06-20 2002-07-25 Haas Beteiligungsgesellschaft Magazine for accommodating tablets and dispensing them individually
US20030190290A1 (en) 2000-06-22 2003-10-09 Calvin Ross Pharmaceutical compositions
CA2417727C (en) 2000-07-31 2009-09-01 Nycomed Danmark A/S Fentanyl composition for nasal administration
DE60120710T2 (en) 2000-08-18 2007-06-14 Pharmacia Corp. FAST CRUMBLING ORAL DRUG PREPARATION CONTAINING VALDECOXIB
US20020026330A1 (en) 2000-08-23 2002-02-28 Klein Edward E. System and method for patient medication management and compliance using a portable computing device
US6417184B1 (en) 2000-09-19 2002-07-09 David M. Ockert Triple drug therapy for the treatment and prevention of acute or chronic pain
GB2368061B (en) 2000-10-20 2005-05-11 Gw Pharmaceuticals Ltd Dose dispensing apparatus
US20050054942A1 (en) 2002-01-22 2005-03-10 Melker Richard J. System and method for therapeutic drug monitoring
US20020160043A1 (en) 2001-02-27 2002-10-31 Dennis Coleman Compositions and method of manufacture for oral dissolvable dosage forms
US20050176790A1 (en) 2001-02-28 2005-08-11 Johannes Bartholomaus Pharmaceutical salts
DE10109763A1 (en) 2001-02-28 2002-09-05 Gruenenthal Gmbh Pharmaceutical salts
CA2439766C (en) 2001-03-15 2008-12-09 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Nebulizer having cooling chamber
JP4850346B2 (en) 2001-03-15 2012-01-11 救急薬品工業株式会社 Mucosal patch
EP1243524A3 (en) 2001-03-16 2004-04-07 Pfizer Products Inc. Pharmaceutical kit for oxygen-sensitive drugs
GB0108215D0 (en) 2001-04-02 2001-05-23 Glaxo Group Ltd Medicament dispenser
GB0108228D0 (en) 2001-04-02 2001-05-23 Glaxo Group Ltd Medicament dispenser
WO2002094172A2 (en) 2001-05-22 2002-11-28 Euro-Celtique Compartmentalized dosage form
US7090830B2 (en) 2001-05-24 2006-08-15 Alexza Pharmaceuticals, Inc. Drug condensation aerosols and kits
NZ529417A (en) 2001-05-24 2006-11-30 Alexza Pharmaceuticals Inc Delivery of alprazolam, estazolam, midazolam or triazolam through an inhalation route
US20030051728A1 (en) 2001-06-05 2003-03-20 Lloyd Peter M. Method and device for delivering a physiologically active compound
US7458374B2 (en) 2002-05-13 2008-12-02 Alexza Pharmaceuticals, Inc. Method and apparatus for vaporizing a compound
JP2005503425A (en) 2001-05-24 2005-02-03 アレックザ モレキュラー デリヴァリー コーポレイション Delivery of drug ester by the prescribed inhalation route
US6564967B1 (en) 2001-06-29 2003-05-20 Brown & Williamson Tobacco Corporation Child resistant tablet dispenser
US20030022910A1 (en) 2001-07-05 2003-01-30 R.T. Alamo Ventures I, Inc. Compositions and methods for sublingual formulations of dihydroergotamine for the treatment of migraine
US6685951B2 (en) 2001-07-05 2004-02-03 R. T. Alamo Ventures I, Inc. Administration of dihydroergotamine as a sublingual spray or aerosol for the treatment of migraine
US20030008005A1 (en) 2001-07-05 2003-01-09 R.T. Alamo Ventures, Inc. Sublingual administration of dihydroergotamine for the treatment of migraine
US20030017175A1 (en) 2001-07-05 2003-01-23 R.T. Alamo Ventures I, Inc. Sublingual administration of dihydroergotamine for the treatment of migraine
US20030068356A1 (en) 2001-07-10 2003-04-10 Pather S. Indiran Sequential drug delivery systems
FR2827517B1 (en) 2001-07-23 2003-10-24 Bioalliance Pharma EXTENDED RELEASE BIOADHESIVE THERAPEUTIC SYSTEMS
EP2244200A3 (en) 2001-08-03 2011-04-27 Hill-Rom Services, Inc. Patient point-of-care computer system
US7144587B2 (en) 2001-08-06 2006-12-05 Euro-Celtique S.A. Pharmaceutical formulation containing opioid agonist, opioid antagonist and bittering agent
US7896192B2 (en) 2001-09-19 2011-03-01 Avancen MOD Corp. Patient controlled timed medication dispenser
US7743923B2 (en) 2001-09-19 2010-06-29 Avanced MOD Corporation Patient controlled timed medication dispenser
US7044302B2 (en) 2001-09-19 2006-05-16 Avancen, Inc. Patient controlled timed oral medication dispenser
BR0212861A (en) 2001-09-26 2004-10-05 Pharmacia Corp Intraorally disintegrating valdecoxib compositions
JP2005506885A (en) 2001-10-22 2005-03-10 スリーエム イノベイティブ プロパティズ カンパニー Transdermal / transmucosal patch packaging
US6863901B2 (en) 2001-11-30 2005-03-08 Collegium Pharmaceutical, Inc. Pharmaceutical composition for compressed annular tablet with molded triturate tablet for both intraoral and oral administration
JP2005516926A (en) 2001-12-17 2005-06-09 アリックス セラピューティクス Analgesic delivery system and method of use
CA2366887C (en) 2001-12-31 2011-11-01 Michael Petersen Replicate incorporating an electronic content monitoring system for use in form-fill-seal applications
ES2563981T3 (en) 2002-02-22 2016-03-17 Gw Pharma Limited Dose delivery system and apparatus
US7204425B2 (en) 2002-03-18 2007-04-17 Precision Dynamics Corporation Enhanced identification appliance
US7074426B2 (en) 2002-03-27 2006-07-11 Frank Kochinke Methods and drug delivery systems for the treatment of orofacial diseases
GB0209782D0 (en) 2002-04-29 2002-06-05 Glaxo Group Ltd Medicament dispenser
US6961541B2 (en) 2002-05-24 2005-11-01 Aeroscout, Inc. Method and apparatus for enhancing security in a wireless network using distance measurement techniques
US6793075B1 (en) 2002-05-30 2004-09-21 Michael Jeter Container for dispensing a liquid and method of using the same
DE10226494A1 (en) 2002-06-14 2004-01-08 Lts Lohmann Therapie-Systeme Ag Film-shaped mucoadhesive dosage forms for administration of cannabis active ingredients
US7500444B2 (en) 2002-06-21 2009-03-10 Glaxo Group Limited Actuation indicator for a dispensing device
JP2004051502A (en) 2002-07-17 2004-02-19 Jcr Pharmaceuticals Co Ltd Stabilized powder preparation containing growth hormone
ATE426460T1 (en) 2002-08-06 2009-04-15 Glaxo Group Ltd DISPENSING DEVICE
US9486581B2 (en) 2002-09-11 2016-11-08 Becton, Dickinson And Company Injector device with force lock-out and injection rate limiting mechanisms
US7295890B2 (en) 2002-09-26 2007-11-13 Stratamed Labs, Inc. Prescription drug compliance monitoring system
US6963289B2 (en) 2002-10-18 2005-11-08 Aeroscout, Ltd. Wireless local area network (WLAN) channel radio-frequency identification (RFID) tag system and method therefor
US6999092B2 (en) 2002-10-25 2006-02-14 Fujitsu Limited Image display apparatus in which a specific area display attribute is modifiable
JP3447009B1 (en) 2002-10-29 2003-09-16 實 平垣 Construct structure and method for producing the same
US6726053B1 (en) 2002-10-29 2004-04-27 John E. Harrold Child resistant multiple dosage blister pack dispenser
GB0226022D0 (en) 2002-11-07 2002-12-18 Corporate Intellectual Propert Device
DE60327883D1 (en) 2002-11-18 2009-07-16 Yaupon Therapeutics Inc ANALGETIC USE OF (S) -NORKETAMINE
WO2004055167A2 (en) 2002-12-13 2004-07-01 Alphavax, Inc. Alphavirus particles and methods for preparation
US7264139B2 (en) 2003-01-14 2007-09-04 Hypoguard Limited Sensor dispensing device
EP1587514B3 (en) 2003-01-31 2011-06-22 Orexo AB A rapid-acting pharmaceutical composition
US20040253307A1 (en) 2003-02-04 2004-12-16 Brian Hague Sugar-free oral transmucosal solid dosage forms and uses thereof
MXPA05008918A (en) 2003-02-24 2006-02-17 Pharmaceutical Productions Inc Transmucosal drug delivery system.
GB0305581D0 (en) 2003-03-11 2003-04-16 Dallas Burston Ltd Dispensing devices
US20060062812A1 (en) 2003-03-11 2006-03-23 Calvin Ross Novel compositions
EP1613324A2 (en) 2003-04-14 2006-01-11 Pain Therapeutics, Inc. Methods for the treatment of pain comprising opioid antagonists
US7276246B2 (en) 2003-05-09 2007-10-02 Cephalon, Inc. Dissolvable backing layer for use with a transmucosal delivery device
US7306812B2 (en) 2003-05-09 2007-12-11 Cephalon, Inc. Dissolvable backing layer for use with a transmucosal delivery device
US6914668B2 (en) 2003-05-14 2005-07-05 International Technologies (Laser) Ltd. Personal identification verification and controlled substance detection and identification system
BR0302017B1 (en) 2003-06-02 2014-10-29 Ems Sigma Pharma Ltda Sublingual pharmaceutical composition based on a benzodiazepine central receptor agonist
US7819847B2 (en) 2003-06-10 2010-10-26 Hewlett-Packard Development Company, L.P. System and methods for administering bioactive compositions
US7118550B2 (en) 2003-07-10 2006-10-10 B. Braun Medical Side-delivery suppository dispenser
US20050049464A1 (en) 2003-08-29 2005-03-03 Lassers Harold Aaron Remote pharmaceutical administration device
US7248165B2 (en) 2003-09-09 2007-07-24 Motorola, Inc. Method and apparatus for multiple frequency RFID tag architecture
US20050065175A1 (en) 2003-09-24 2005-03-24 Xanodyne Pharmacal, Inc. Oral transmucosal methadone
US7268109B2 (en) 2003-10-02 2007-09-11 Elan Pharmaceuticals, Inc. Method for reducing pain
US8157788B2 (en) 2003-11-06 2012-04-17 Paolo L. Manfredi Multi-site drug delivery platform
US7540998B2 (en) 2003-11-19 2009-06-02 Biocompatibles Uk Limited Delivery system and prescription method for interstitial radiation therapy using enhanced parametric release sterilization techniques
US7780973B2 (en) 2003-12-15 2010-08-24 Ethicon Endo-Surgery, Inc. Method and device for minimally invasive implantation of biomaterial
US6999028B2 (en) 2003-12-23 2006-02-14 3M Innovative Properties Company Ultra high frequency radio frequency identification tag
CA2549642C (en) 2003-12-31 2012-10-30 Cima Labs Inc. Effervescent oral opiate dosage forms and methods of administering opiates
WO2005065317A2 (en) 2003-12-31 2005-07-21 Cima Labs Inc. Effervescent oral fentanyl dosage form
EA010826B1 (en) 2003-12-31 2008-12-30 Сайма Лэбс Инк. Fentanyl dosage form for oral administration, methods of making thereof and methods of treatment
US20070036853A1 (en) 2003-12-31 2007-02-15 Cima Labs Inc. Generally linear effervescent oral fentanyl dosage form and methods of administering
US20050150489A1 (en) 2004-01-12 2005-07-14 Steve Dunfield Dispensing medicaments based on rates of medicament action
AU2005209199B2 (en) 2004-01-16 2008-09-11 Biodel Inc. Sublingual drug delivery device
BRPI0508769A (en) 2004-03-30 2007-08-28 Euro Celtique Sa tamper-resistant dosage form comprising an adsorbent and an adverse agent
US20050226923A1 (en) 2004-04-07 2005-10-13 Gassert Chad M Venlafaxine compositions in the form of microtablets
US7004111B2 (en) 2004-05-26 2006-02-28 Keryl Olson Treat dispenser
US20060026035A1 (en) 2004-07-28 2006-02-02 William Younkes Computer aided interactive medical management information and control system and method
US20060045865A1 (en) 2004-08-27 2006-03-02 Spherics, Inc. Controlled regional oral delivery
GB0420016D0 (en) 2004-09-09 2004-10-13 Leuven K U Res & Dev Controlled release oral delivery system
US20060067978A1 (en) 2004-09-29 2006-03-30 Bausch & Lomb Incorporated Process for preparing poly(vinyl alcohol) drug delivery devices
US7119690B2 (en) 2004-10-08 2006-10-10 Proximities, Inc. Identification band using serpentine paths to detect tampering
US7388493B2 (en) 2004-10-08 2008-06-17 Bartronics America, Inc. Method and system for preventing unauthorized removal and use of an RFID apparatus
CA2584606A1 (en) 2004-10-21 2006-05-04 Bayer Healthcare Llc Sensor-dispensing device and mechanism for extracting sensor
CN2776369Y (en) 2004-10-22 2006-05-03 瓦索金爱尔兰有限公司 Distribution system
US20060089858A1 (en) 2004-10-25 2006-04-27 Tun Ling Means and method of applying RFID and PKI technologies for patient safety
FR2883179B1 (en) 2005-03-18 2009-04-17 Ethypharm Sa COATED TABLET
CN101151021A (en) 2005-03-28 2008-03-26 奥瑞克索股份公司 New pharmaceutical compositions useful in the treatment of pain
US20070020186A1 (en) 2005-07-22 2007-01-25 Alpex Pharma S.A. Solid dosage formulations of narcotic drugs having improved buccal adsorption
WO2007035913A2 (en) 2005-09-21 2007-03-29 Kurve Technology, Inc. Medicament delivery control, monitoring, and reporting system and method
US20070104763A1 (en) 2005-11-10 2007-05-10 Navinta Llc Composition of fentanyl citrate oral solid transmucosal dosage form, excipient and binding material therefore, and methods of making
JP2009522288A (en) 2005-12-28 2009-06-11 アルザ コーポレイション Stable therapeutic dosage form
US9066847B2 (en) 2007-01-05 2015-06-30 Aceirx Pharmaceuticals, Inc. Storage and dispensing devices for administration of oral transmucosal dosage forms
US8202535B2 (en) 2006-01-06 2012-06-19 Acelrx Pharmaceuticals, Inc. Small-volume oral transmucosal dosage forms
US8753308B2 (en) 2006-01-06 2014-06-17 Acelrx Pharmaceuticals, Inc. Methods for administering small volume oral transmucosal dosage forms using a dispensing device
US8535714B2 (en) 2006-01-06 2013-09-17 Acelrx Pharmaceuticals, Inc. Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain
US8252329B2 (en) 2007-01-05 2012-08-28 Acelrx Pharmaceuticals, Inc. Bioadhesive drug formulations for oral transmucosal delivery
US8252328B2 (en) 2006-01-06 2012-08-28 Acelrx Pharmaceuticals, Inc. Bioadhesive drug formulations for oral transmucosal delivery
US8357114B2 (en) 2006-01-06 2013-01-22 Acelrx Pharmaceuticals, Inc. Drug dispensing device with flexible push rod
US8865743B2 (en) 2006-01-06 2014-10-21 Acelrx Pharmaceuticals, Inc. Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain
US9289583B2 (en) 2006-01-06 2016-03-22 Acelrx Pharmaceuticals, Inc. Methods for administering small volume oral transmucosal dosage forms using a dispensing device
WO2008085764A1 (en) 2007-01-05 2008-07-17 Acelrx Pharmaceuticals, Inc. Storage and dispensing devices for administration of oral transmucosal dosage forms
WO2007084290A2 (en) 2006-01-12 2007-07-26 Orexigen Therapeutics, Inc. Compositions of an anticonvulsant and psychotherapeutic and methods of using the same for reversing weight gain
US20070190130A1 (en) 2006-02-16 2007-08-16 Mark William A Protein hydrolysate excipients
US20070260491A1 (en) 2006-05-08 2007-11-08 Pamela Palmer System for delivery and monitoring of administration of controlled substances
US7581657B2 (en) 2006-05-15 2009-09-01 Dickmann Alexander J Bandage dispenser
US20070286900A1 (en) 2006-06-09 2007-12-13 Catherine Herry Low dose tablets of opioid analgesics and preparation process
US20070299687A1 (en) 2006-06-23 2007-12-27 Pamela Palmer Inpatient system for patient-controlled delivery of oral transmucosal medications dosed as needed
US7744558B2 (en) 2007-06-06 2010-06-29 Maag R John Pill plunger
US20090010992A1 (en) 2007-07-03 2009-01-08 Aceirx Pharmaceuticals, Inc. Drug formulations for oral transmucosal delivery to pediatric patients
JP5632284B2 (en) 2007-08-07 2014-11-26 エーセルアールエックス ファーマシューティカルズ, インコーポレイテッド Compositions and methods for sedation and analgesia during treatment using oral transmucosal dosage forms
US8945592B2 (en) 2008-11-21 2015-02-03 Acelrx Pharmaceuticals, Inc. Sufentanil solid dosage forms comprising oxygen scavengers and methods of using the same
WO2010107761A1 (en) 2009-03-18 2010-09-23 Acelrx Pharmaceuticals, Inc. Improved storage and dispensing devices for administration of oral transmucosal dosage forms
US8062248B2 (en) 2009-07-14 2011-11-22 Charles Edward Kindel Pill dispenser for pets
US20110091544A1 (en) 2009-10-16 2011-04-21 Acelrx Pharmaceuticals, Inc. Compositions and Methods for Mild Sedation, Anxiolysis and Analgesia in the Procedural Setting
US20110288128A1 (en) 2010-05-21 2011-11-24 Acelrx Pharmaceuticals, Inc. Oral Transmucosal Administration of Sufentanil

Also Published As

Publication number Publication date
US10507180B2 (en) 2019-12-17
US8865743B2 (en) 2014-10-21
US20180161270A1 (en) 2018-06-14
EP2114383B1 (en) 2010-07-21
US20160213606A1 (en) 2016-07-28
US20190125659A1 (en) 2019-05-02
CA2673856C (en) 2018-11-27
WO2008085765A3 (en) 2008-11-27
US9320710B2 (en) 2016-04-26
US10245228B2 (en) 2019-04-02
DE602007007991D1 (en) 2010-09-02
US20080268023A1 (en) 2008-10-30
WO2008085765A2 (en) 2008-07-17
KR101545754B1 (en) 2015-08-19
KR20090099009A (en) 2009-09-18
US9744129B2 (en) 2017-08-29
ATE474564T1 (en) 2010-08-15
US20150105424A1 (en) 2015-04-16
JP2010532745A (en) 2010-10-14
PT2114383E (en) 2010-08-16
EP2114383A2 (en) 2009-11-11
DK2114383T3 (en) 2010-10-04

Similar Documents

Publication Publication Date Title
US10507180B2 (en) Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain
US8778393B2 (en) Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain
US10709881B2 (en) Apparatus for administering small volume oral transmucosal dosage forms
CA2673837C (en) Storage and dispensing devices for administration of oral transmucosal dosage forms
US9289583B2 (en) Methods for administering small volume oral transmucosal dosage forms using a dispensing device
CA2673880C (en) Storage and dispensing devices for administration of oral transmucosal dosage forms
CN101621994B (en) For treating a small amount of oral transmucosal dosage forms containing sufentanil of pain

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20121220