CA2707980C - Misuse preventative, controlled release formulation - Google Patents

Misuse preventative, controlled release formulation Download PDF

Info

Publication number
CA2707980C
CA2707980C CA2707980A CA2707980A CA2707980C CA 2707980 C CA2707980 C CA 2707980C CA 2707980 A CA2707980 A CA 2707980A CA 2707980 A CA2707980 A CA 2707980A CA 2707980 C CA2707980 C CA 2707980C
Authority
CA
Canada
Prior art keywords
formulation
microparticles
controlled release
core
cellulose
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.)
Active
Application number
CA2707980A
Other languages
French (fr)
Other versions
CA2707980A1 (en
Inventor
Miloud Rahmouni
Vinayak Sant
Sonia Gervais
Damon Smith
Frederic Duffayet
Shams Rustoms
Ali El-Jammal
Bobby-Ernst Boursiquot
Ali Bichara
Jean-Michel Ndong
Angela Ferrada
Fouzia Soulhi
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.)
Paladin Labs Inc
Endo Ventures Ltd
Original Assignee
Paladin Labs Barbados Inc
Paladin Labs Inc
Paladin Labs Europe Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Paladin Labs Barbados Inc, Paladin Labs Inc, Paladin Labs Europe Ltd filed Critical Paladin Labs Barbados Inc
Publication of CA2707980A1 publication Critical patent/CA2707980A1/en
Application granted granted Critical
Publication of CA2707980C publication Critical patent/CA2707980C/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
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • A61K9/2081Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets with microcapsules or coated microparticles according to A61K9/50
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • 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
    • 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
    • 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
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • 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
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • 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
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • 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
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • 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
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2086Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
    • A61K9/209Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat containing drug in at least two layers or in the core and in at least one outer layer
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2813Inorganic 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/282Organic compounds, e.g. fats
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • A61K9/2846Poly(meth)acrylates
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • A61K9/2866Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

Disclosed is a misuse preventative, controlled release formulation comprising a core comprising a superabsorbent material (for example, polycarbophil), a controlled release coat surrounding the core, and a plurality of controlled release microparticles having a pharmaceutically active agent (for example, an opioid analgesic) disposed within the core, the coat, or both the core and the coat.
When crushed, either intentionally or accidentally, and exposed to an aqueous medium, the superabsorbent material present in the core swells to encapsulate the microparticles, which remain substantially intact thereby retarding the release of the pharmaceutically active agent from the formulation. Also disclosed is a method of using the misuse preventative, controlled release formulation to deliver a pharmaceutically active agent to a mammal, for example, a human, in need thereof.

Description

MISUSE PREVENTATIVE, CONTROLLED RELEASE FORMULATION
TECHNICAL FIELD
10001] The present invention relates generally to a controlled release formulation for the delivery of at least one pharmaceutically active agent, and more specifically, the invention relates to a misuse preventative, controlled release formulation, which maintains its controlled release properties for at least one pharmaceutically active agent even when bisected or crushed and exposed to various media.
BACKGROUND ART
[0002] Although significant developments have been made in the field of drug delivery, concerns remain for drugs (for example, opioid analgesics) that are subject to abuse.
Furthermore, the numbers of legitimate patients misusing such drugs, either deliberately or accidentally, represents a serious medical problem. In particular, patient risk can be heightened when controlled release formulations are used because larger amounts of the pharmaceutically active agent typically are incorporated into these formulations to facilitate reduced dosing-frequency. However, while controlled release formulations may offer greater convenience and an improved adverse event profile, serious problems can occur if the control release mechanism is compromised in any way, for example, by accidental chewing or grinding of, or other damage to, the tablet, or co-ingestion with alcohol. Under these scenarios, immediate release of the pharmaceutically active agent followed by rapid absorption of up to a total daily dose of the pharmaceutical agent can have potentially fatal consequences.
[0003] While a number of approaches have been tried to address the abuse and misuse of certain drugs, no effective approach has yet been commercialized. To date, the approaches employed include, for example, deterrent formulations, agonist/antagonist formulations, and pro drug formulations.
[00041 Deterrent formulations are formulations that contain a noxious substance, such as, capsaicin, an emetic, or niacin. The aim is to prevent deliberate abuse by inflicting a painful or otherwise unpleasant reaction should the formulation be crushed or otherwise damaged prior to ingestion. For example, U.S. Patent Publication Nos 2003/0068370, 2003/0068392 and 2007/0020188 describe incorporation of aversive agents (e.g., a bitter agent, an irritant, or an
4 emetic agent) into a dosage containing an opioid analgesic. The aversive agents discourage an abuser from tampering with the dosage form and thereafter inhaling or injecting the tampered dosage. The potential risk of such additives to the legitimate user who accidentally damages the tablet is not addressed by such formulations.
[0005] Antagonist formulations contain inhibitors (antagonists) of the therapeutic drug. When the formulation is crushed, the inhibitors are intended to prohibit or reverse the action of the pharmaceutically active agent thereby reducing or eliminating any benefit for non-medical use.
For example, naloxone is combined with pentazocine (Talwin D, sold by Sanofi-Winthrop) to deter parenteral abuse of pentazocine. Naloxone is intended to block the binding of pentazocine to opioid receptors. Similarly, naloxone is added to a buprenorphine-containing formulation (Temgesic8, sold by Reckitt & Colman). It is understood, however, that this approach, can expose legitimate patients to unnecessary drugs, and can potentially inhibit effective therapy because the inhibitors may be released during normal passage through the gastrointestinal tract.
These formulations also assume that effective inhibition can be achieved (i.e., that the bio availability, pharmacokinetics and relative affinities of the agonist and antagonist can be matched so as to elicit effective inhibition in the intended recipient). U.S.
Patent Nos. 3,773,955 and 3,966,940, for example, describe formulations containing combinations of opioid agonists and antagonists, in which the antagonist does not block the therapeutic effect when the mixture is administered orally but blocks analgesia, euphoria or physical dependence when administered parenterally in a crushed form by an abuser.
[00061 Prodrug formulations rely on in vivo metabolic conversion of the prodrug into the active drag by enzymes found, for example, in the gastrointestinal tract. While these formulations may prevent euphoria via intravenous or nasal administration of the drug, they do not address the problems associated with potential intoxication (for example, alcohol intoxication) post oral administration.
[00071 Because of such limitations with existing technologies, there exists an ongoing need for misuse preventative, controlled release formulations that can reduce the risk of intentional abuse and accidental misuse of formulations containing a pharmaceutically active agent.

DISCLOSURE OF THE INVENTION
[0008] The invention is based, in part, upon the discovery that it is possible to create a drug delivery platform that permits the controlled release of a pharmaceutically active agent disposed within the formulation even after being sectioned (for example, bisected) or crushed. The platform is particularly useful for the administration of pharmaceutically active agents that are capable of abuse and/or that have a narrow therapeutic index Agents capable of abuse, include, for example, analgesics (for example, opioid analgesics), hypnotic agents, anxiolytic agents, central nervous system (CNS) and respiratory stimulating agents, and agents having a narrow therapeutic index.
[0009] In one aspect, the invention provides a controlled release formulation, comprising: (a) a core comprising a superabsorbent material (for example, polycarbophil), (b) a controlled release coat surrounding the core; and (c) a plurality of controlled release microparticles having a pharmaceutically active agent disposed therein, wherein the microparticles are disposed within the core, the coat, or both the core and the coat. As a result, the formulations are designed to have two controlled release mechanisms (the coat and the microparticles), which work together in an intact formulation. However, when crushed to compromise the coating, the microparticles remain substantially intact to control the release of the pharmaceutically active agent and prevent dose dumping, [0010] If exposed to an aqueous environment, at least one pharmaceutically active agent is released from the intact formulation over a prolonged period of time (for example, for at least 6 hours, at least 8 hours, at least 12 hours, at least 18 hours, or at least 24 hours). In certain embodiments, at least 50 %, preferably 60 %, more preferably 70 %, and even more preferably 80 % of at least one pharmaceutically active agent is prevented from being released substantially immediately (for example, within 30 minutes) from the intact formulation.
[0011] If the formulation is crushed to break the controlled release coat and expose the core, and then exposed to an aqueous environment, the superabsorbent material swells to create a hard, rigid gel that traps the microparticles, which remain substantially intact.
The hard gel creates an unpleasant experience if the crushed formulation is snorted up the nose and absorbs the nasal secretions that would otherwise permit absorption via this route. Furthermore, once the hard gel has formed following exposure to an extraction media, the residting gel cannot be pushed through a needle of a syringe. Although the controlled release properties of the coating may be compromised by crushing, the microparticles still permit the controlled release of the pharmaceutically active agent and prevent the agent from being released substantially immediately from the formulation. In certain embodiments, at least 50 %, preferably 60 %, more preferably 70 %, and even more preferably 80 % of at least one pharmaceutically active agent is prevented from being released substantially immediately (for example, within 30 minutes) from the formulation. As a result, the formulations of the invention reduce or eliminate the potential for dose dumping in water, alcohol (for example, ethanol), and other media of various pH even if the formulations have been broken or crushed.
[0012] It is understood that in certain embodiments, the controlled release microparticles can be disposed within the core or the coat. In other embodiments, the controlled release microparticles (which can be the same or different) are disposed within both the core and the coat. It is understood that the choice of location of the particles will depend upon the release profile desired for the formulation. For example, if release over 8 hours is desired, then the particles may be located within the coat. On the other hand, if release over 24 hours is desired, then the particles may be located within the core, or within both the core and the coat.
[0013] In certain embodiments, the core is monolithic. The monolithic core optionally comprises microparticles disposed therein. It is understood, however, that under certain circumstances the core can comprise a plurality of different release matrices, which can be, for example, in the form of a bilayer or a multilayer that contains two, three or more layers. In one bilayer embodiment, a first layer contains the drug containing microparticles and a second layer contains free drug (i.e., drug not present in or associated with microparticles). As a result, drug is released faster from the second layer that lacks the microparticles than from the first layer that contains the microparticles. Furthermore, it is contemplated that, depending upon the desired release profiles, one layer of the bilayer can contain one set of microparticles having one set of release kinetics and the other layer of the bilayer can contain a second, different set of microparticles having a second, different set of release kinetics.
[0014] In certain embodiments, the superabsorbent material is present such that it constitutes about 10 % (w/w) to about 70 % (w/w) of the core. In other embodiments, the superabsorbent material constitutes about 30 % (w/w) to about 50 % (w/w) of the core. In addition, relative to the intact formulation, in certain embodiments, the volume of the core constitutes about 5 % to about 40 % of the intact formulation, about 10 % to about 30 % of the intact formulation, or about 15 % to about 20 % of the intact formulation. In certain embodiments, the volume of the core constitutes at least 30 %, at least 20 %, or at least 15% of the final volume of the resulting intact formulation.
(00151 The controlled release microparticles comprise a controlled release medium (for example, cross-linked high amylase starch sold under the Tradename CONTR.AMIL:0) from Labopharm, Inc., Laval, Canada) that controls the release of the pharmaceutically active agent disposed therein and/or a controlled release coating or film. The microparticles have an average diameter in the range from about 1 m to about 1000 p.im. The microparticles, due to their small size and high radius of curvature, resist crushing when the formulation is crushed, for example, with a conventional pill crusher or between spoons or in a pestle and mortar. In one embodiment, the micropartieles have an average diameter in the range from about of 200 am to about 900 urn, or from about 300 am to about 800 um. The microparticles under certain circumstances have an average diameter of about 700 um. In another embodiment, the controlled release microparticles have an average diameter in the range of from about 1 p,m to about 400 um, from about 5 am to about 300 am, or from about 10 um to about 200 um. The microparticles can have an average diameter of about 100 am.
[0016] In addition, it is understood that the formulations can contain microparticles that contain the same pharmaceutically active agent or the same combination of two or more pharmaceutically active agents. Alternatively, the formulations can contain microparticles where one population of microparticles contain one agent and another population of microparticles contain a second, different agent.
[0017] In another aspect, the invention provides a method of providing controlled release of a pharmaceutically active agent to a mammal, for example, a human. The method comprises orally administering to an individual in need of the pharmaceutically active agent one Or more of the controlled release formulations described herein.

In one aspect, there is provided a controlled release formulation, comprising:
(a) a core comprising a superabsorbent material selected from the group consisting of polycarbophil, polycarbophilic calcium, polymethacrylic acid, polyacrylic acid, and a mixture thereof, the superabsorbent material comprising from 10% to 70% (w/w) of the core; (b) a controlled release coat surrounding the core; and (c) a plurality of controlled release microparticles having a pharmaceutically active agent disposed therein, wherein the microparticles particles are disposed within the core, the coat, or both the core and the coat, wherein the formulation has a hardness from about 200 N to about 400 N, and wherein the formulation, and wherein the formulation (i) when intact and exposed to an aqueous environment, the pharmaceutically active agent is released from the formulation over a prolonged period of time, and (ii) when crushed to break the controlled release coat and expose the core, and exposed to an aqueous environment, the superabsorbent material swells to create a hard gel that traps the microparticles, and the microparticles provide controlled release of the pharmaceutically active agent.
In another aspect, there is provided a controlled release formulation, comprising:
(a) a core comprising from about 10 % to about 70% (w/w) polycarbophil, polycarbophilic calcium, or a mixture thereof; (b) a controlled release coat surrounding the core; and (c) a plurality of controlled release microparticles having a pharmaceutically active agent disposed therein, wherein the microparticles particles are disposed within the core, the coat, or both the core and the coat, wherein the formulation has a hardness from about 200 N to about 400 N, and wherein the formulation (i) when intact and exposed to an aqueous environment, the pharmaceutically active agent is released from the formulation over a prolonged period of time, and (ii) when crushed to break the controlled release coat and expose the core, and exposed to an aqueous environment, the superabsotbent material swells to create a hard gel that traps the microparticles, and the microparticles provide controlled release of the pharmaceutically active agent.
5a BRIEF DESCRIPTION OF THE DRAWINGS
[0018] The foregoing and other objects, features and advantages of the invention will become apparent from the following description of preferred embodiments, as illustrated in the accompanying drawings. Like referenced elements identify common features in the corresponding drawings, The drawings are not necessarily to scale, with emphasis instead being placed on illustrating the principles of the present invention, in which:
[0019] FIGURE 1 shows a schematic representation of exemplary misuse preventative, controlled release formulations where controlled release micropartieles containing an agent of interest are disposed within the coat (FIGURES IA and ID), the core (FIGURES
1B and 1E), or within both the core and the coat (FIGURES IC and 1F), wherein, in FIGURES
IA-1C, the core is monolithic and in FIGURES 1D-1F, the core is a bilayer;
[0020] FIGURE 2 is a graph showing the in vitro dissolution profile of Tramadol 1-ICI from an intact, exemplary controlled release formulation of the invention in a U.S.P.
Type I Apparatus in phosphate buffer pH 6.8;
[0021] FIGURE 3 is a graph showing the in vitro dissolution profile of Tramadol HC1 from an intact, exemplary controlled release formulation of the invention in a U.S.P.
Type I Apparatus where the solvent is water (-o-), 20% ethanol 40% ethanol (-A-), 60%
ethanol (- f -), 80%
ethanol (-0-) or 100% ethanol (-=-);
[0022] FIGURE 4 is a graph showing the in vitro dissolution profile of Tramadol HC1 from an intact, exemplary controlled release formulation of the invention in a U.S.P.
Type I Apparatus as a function of pH where the solvent is water (-s-), buffer at pH 1.2 (-r-), buffer at pH 3.0 (-0-), buffer at pH 5.0 (-v-), or buffer at pH 6.8 (-6,-);
[0023] FIGURE 5 is a graph showing the in vitro dissolution profile of Tramadol HC1 from an intact, exemplary controlled release tablet of the invention (-41-) or from half a tablet (a bisected tablet) of the invention where the release values have been normalized relative to the intact tablet (-m-) using a U.S.P. Type I Apparatus with phosphate buffer pH 6.8;
[0024] FIGURE 6 is a photograph showing five vials, where the first vial contains 2 mL of water and the second through the fifth vials (inverted) contain different tablets of controlled release formulations of the invention each of which had been crushed in a pill crusher and
6 exposed to 2 mL of water to produce a hard gel that remained at the bottom of each vial even when inverted;
[0025] FIGURE 7 is a photograph showing seven inverted vials each containing crushed tablets of the invention that had been exposed to 10 rnL of (i) water at room temperature for 15 minutes with agitation (vial 1), (ii) water at 50 C for 15 minutes with agitation (vial 2), (iii) water at 75 C
for 15 minutes with agitation (vial 3), (iv) water at 100 C for 15 minutes with agitation (vial 4), (v) acidic media (pH 1.2) at room temperature for 15 minutes with agitation (vial 5), (vi) basic media (pH 7.5) at room temperature for 15 minutes with agitation (vial 6), and (vii) 40 % ethanol in water at room temperature for 15 minutes with agitation (vial 7);
[0026] FIGURE 8 is a bar chart showing the effect of different ethanol concentrations on Tramadol release from crushed tablets of the invention (bars with light shading) or Ultrarn ER
(bar in dark shading) after incubation in 900 mL of extraction media for 30 minutes at 37 C in a U.S.P. Type I Apparatus;
[0027] FIGURE 9 is a bar chart showing the effect of pH on Tramadol release from crushed tablets of the invention (bars with light shading) or Ultram ER (bar in dark shading) after incubation in 900 inL of extraction media of various pH for 30 minutes at 37 C
in a U.S.P. Type 1 Apparatus.
[0028] FIGURES 10A and 10B are graphs showing the mean plasma concentration of Tramadol released from an exemplary 100 mg tablet following single-dose administration to adult humans under fasting conditions (FIGURE 10A) or under fed conditions (FIGURE 10B);
[0029] FIGURE HA AND 11B are graphs showing the in vitro dissolution profiles of an embodiment containing 40 mg oxycodone HC1 in a U.S.P. Type I Apparatus at 100 rpm for twelve hours from either an intact tablet (FIGURE 11A) or a crushed tablet (FIGURE 11B) in phosphate buffer pH 6.8 (-0-) or buffer containing 40% ethanol (-o-);
[0030] FIGURE 12 is a graph showing the in vitro dissolution profiles of an embodiment containing 40 mg oxycodone HC1 in a U.S P. Type I Apparatus at 100 rpm for twelve hours from either an intact tablet in phosphate buffer pH 6.8 (-=-), or a bisected tablet in phosphate buffer pH 6.8 (-A.-);
7 [0031] FIGURE 13 is a graph showing the in vitro dissolution profile of a coated biloer embodiment containing 20 mg oxycodone HCl/650 mg acetaminophen, where the release of oxycodone was measured in a U.S.P. Type I Apparatus at 100 rpm in acid at pH
1.2 for 1 hour and then in a phosphate buffer at pH 6.8 for 11 hours;
[0032] FIGURES 14A and 14B are graphs showing the in vitro dissolution profiles of an embodiment containing 150 mg Tramadol HC1 in a U.S.P. Type I Apparatus at 100 rpm in phosphate buffer pH 6.8 from three different lots of intact tablets (FIGURE
14A) or from crushed tablets (FIGURE 14B);
(0033] FIGURES 15A and 15B are graphs showing the in vitro dissolution profiles of an embodiment containing 150 mg Tramadol HC1 in a U.S.P. Type I Apparatus at 100 rpm in water containing 60 % ethanol from three different lots of intact tablets (FIGURE
15A) or from crushed tablets (FIGURE 15B);
[0034] FIGURES 16A and 1613 are graphs showing the in vitro dissolution profiles of an embodiment containing 200 mg Trarnadol 1-ICI in a U.S.P. Type I Apparatus at 100 rpm in phosphate buffer pH 6.8 or water from either intact tablets (FIGURE 16A) or from crushed tablets (FIGURE 16B);
[0035] FIGURES 17A and 17B are graphs showing the in vitro dissolution profiles of an embodiment containing 200 mg Tramadol HC1 in a U.S.P. Type I Apparatus at 100 rpm in phosphate buffer pH 6.8 alone (-*-) or water containing 20 % ethanol (-A-), 40 % ethanol (-E-), or 60 % ethanol (- = -) either from intact tablets (FIGURE 17A) or from crushed tablets (FIGURE 17B); and (0036] FIGURES 18A and 18B are graphs showing the in vitro dissolution profiles of an embodiment containing 30 mg hydrocodone in a U.S.P. Type I Apparatus at 100 rpm in phosphate buffer pH 6.8 (-s-) or acid pH 1.2 (- A- ) either from intact tablets (FIGURE 18A) or from crushed tablets (FIGURE 18B) BEST MODES OF CARRYING OUT THE MENTION
10037] The invention is based, in part, upon the discovery that it is possible to produce a controlled release platform that provides pharmaceutical formulations less susceptible to intentional abuse and accidental misuse than other controlled release formulations and is free
8 from noxious additives, active ingredient antagonists, prodrugs, and the like.
The formulations maintain their controlled release properties when sectioned (for example, bisected) as can happen, for example, when a subject breaks a tablet in half to make it easier to swallow.
Furthermore, even when crushed, the formulations of the invention prevent dose damping -- because the microparticles contained within the formulation remain substantially intact and retain their controlled release properties.
[0038] The invention provides a controlled release formulation, comprising:
(a) a core comprising a superabsorbent material (for example, polycarbophil); (b) a controlled release coat surrounding the core; and (c) a plurality of controlled release microparticles having a -- pharmaceutically active agent disposed therein, wherein the mieroparticles are disposed within the core, the coat, or both the core and the coat. The formulations have two controlled release mechanisms (the coat and the microparticles), which work together in an intact formulation.
However, even when crushed to compromise the coating, the microparticles remain substantially intact to control the release of the pharmaceutically active agent and prevent dose dumping. As -- used herein, the term "dose dumping" is understood to mean an uncontrolled release of a pharmaceutically active agent where at least 80% of the pharmaceutically active agent in the formulation is released within 30 minutes (a specification that can be used to characterize a formulation as an immediate release formulation).
[0039] Figure 1 shows certain embodiments (Figures 1A-1F) of the formulation of the -- invention. Each formulation 10 contains a core 20 and a coat 30. In Figures 1A and 1D, formulation 10 contains controlled release microparticles 40 located within coat 30. In Figures 1B and 1E, formulation 10 contains controlled release microparticles 40 located within core 20.
In Figures 1C and 1F, formulation 10 contains controlled release microparticles 40 located within both core 20 and coat 30. In Figures 1A-1C, the core is monolithic. In Figures 11)-1F, -- the core is shown to be a bilayer having a first layer 50 and a second, different layer 60. It is understood, however, that the core can comprise a multilayer having two or more (for example, three, four or more) layers of different materials. In each of the embodiments shown in Figure 1, the microparticles control the release of the active ingredient irrespective of whether the tablet is intact or compromised (for example, by bisection or crushing).
9 [0040] Under normal use, coat 30 protects core 20 from exposure to the solvent thereby preventing the swelling of the superabsorbent material in the core. As a result, the pharmaceutically active agent is released steadily from the formulation. If drug containing controlled release microparticles are located within coat 30, then the drug is released from coat 30 as the solvent permeates the coat. If drug containing controlled release microparticles are located within both coat 30 and core 20, then the drug initially is released from the microparticles in the coat. Over time, as the solvent gradually permeates through the coat and then accesses core 20, the drug is released from the microparticles located within the core. The formulations are designed so that coat 30 maintains sufficient integrity (for example, the coat acts like a rigid or semi-rigid net) such that the superabsorbent material in core 20 is prevented from swelling and disrupting the integrity of the tablet.
[0041] It is contemplated that the compositions described herein can be used for the delivery of one or more (for example, two, three, four or more) pharmaceutically active agents. For example, the micropaitieles disposed in the core can contain a first pharmaceutically active agent and microparticles disposed in the coat can contain a second, different pharmaceutically active agent. Alternatively, the microparticles disposed in the core and/or the coat can contain two or more different pharmaceutically active agents. Furthermore, it is contemplated that the core and/or the coat can comprise two or more different populations of microspheres where each population contains the same or a different pharmaceutically active agent. It is understood that the excipients present in each layer may vary. Furthermore, depending upon the release kinetics desired, a pharmaceutically active agent can be disposed in the core and/or the coat but not present within the microparticles. For example, a first pharmaceutically active agent disposed within microparticles can be present in the coat but the same or different pharmaceutically active agent not present in microparticles can be present in the core. Conversely, a first pharmaceutically active agent not present in microparticles can be present in coat but the same or different pharmaceutically active agent disposed in microparticles can be present in the core.
[0042] In certain embodiments, the core is monolithic (see, Figures 1A-1C).
The monolithic core optionally can comprise microparticles disposed therein. It is understood, however, that under certain circumstances the core can comprise a plurality of different release matrices, which can be, for example, in the form of a bilayer or a multilayer that contains two, three or more layers (see, Figures 1D-1F). One of the layers can act can as an immediate release layer and another layer can act as a controlled release layer. Alternatively, at least two of the layers can have controlled release properties. In one embodiment, one layer can release one pharmaceutically active agent and another layer can release a different pharmaceutically active agent, which can be released at the same or at different rates. In another embodiment, one layer can release one pharmaceutically active agent at one rate and another layer can release the same pharmaceutically active agent at a different rate (i.e., faster or slower than the first layer). In one bilayer embodiment, a first layer contains drug containing microparticles and a second layer contains free drug (i_e , not contained within in or associated with microparticles). As a result, the drug is released faster from the second layer that lacks the microparticles than from the first layer that contains the microparticles. Furthermore, it is contemplated that, depending upon the desired release profiles, one layer of the bilayer can contain one population of microparticles having one set of first release kinetics and the other layer of the bilayer can contain a second, different population of microparticles having a second, different set of release kinetics.
[0043] In the case of an intact formulation, when exposed to an aqueous environment (for example, a solution containing at least 10 % (v/v) water), at least one pharmaceutically active agent is released from the intact formulation over a prolonged period of time (for example, for at least 8 hours, at least 12 hours, at least 18 hours, or at least 24 hours). In certain embodiments, at least 50 %, preferably 60 %, more preferably 70 %, and even more preferably 80 % of at least one pharmaceutically active agent is prevented from being released substantially immediately (for example, within 30 minutes) from the formulation when exposed to an extraction medium, for example, water, aqueous solutions ranging in pH from 1.2 to 6.8, and different ethanolic media (for example, water containing 20 % ethanol, 40 % ethanol, 60 % ethanol, or 80 % ethanol and 100 % ethanol). These features are shown, for example, in Figures 2-4, which are discussed in more detail in Example 2.
[0044] When the formulation is bisected, for example, axially bisected, as can happen when a patient breaks a tablet in half to make it easier to swallow, the controlled release coating becomes compromised. However, the combination of the residual coat surrounding the core, partial swelling of the core and the controlled release properties of the microparticles permit the formulations to have a release profile of the pharmaceutically active agent substantially the same as the intact tablet. Furthermore, even when bisected, the formulations of the invention permit the release of the pharmaceutically active agent over at least 12 hours, at least 18 hours, or over at least 24 hours. In certain embodiments, at least 50 %, preferably 60 %, more preferably 70 %, and even more preferably 80 % of at least one pharmaceutically active agent is prevented from being released substantially immediately (for example, within 30 minutes) from the formulation - when exposed to an extraction medium, for example, water, aqueous solutions ranging in pH
from 1.2 to 6.8, and different ethanolic media (for example, water containing 20 % ethanol, 40 %
ethanol, 60 % ethanol, or 80 % ethanol and 100 % ethanol). These features are shown, for example, in Figure 5 and in Figure 12.
[00451 When the formulation is crushed (for example, with a commercially available pill crusher to break formulation into at least 10 particles or more) to break the controlled release coat and expose the core, and then exposed to an aqueous environment, the superabsorbent material swells rapidly (for example, within about 30 seconds) to create a hard gel that traps the microparticles. eased in part upon their small size (high radius of curvature), the rnicroparticles resist the crushing process and remain substantially intact. The hard gel provides an unpleasant experience if the crushed formulation is snorted up a nostril and gel formation occurs within the nostril. This process has the advantage that the nasal secretions needed for absorption of the active ingredient into the blood-stream are absorbed by the superabsorbent material preventing intoxication via this route, Similarly, if the formulation is crushed and exposed to an aqueous environment to extract the pharmaceutically active agent, the superabsorbant material in the core can absorb the extraction medium leaving little or no extraction medium to administer (see, Figures 6 and 7, which are discussed in Example 4). In addition, the hard gel that is formed during this process is difficult to draw or push though a syringe needle.
[0046] Although the controlled release properties of the coating are compromised by crushing, the microparticles still permit the controlled release of the pharmaceutically active agent and prevent the agent from being released substantially immediately from the formulation (i.e., the microparticles provide controlled release of the pharmaceutically active agent). For example, at least 50 %, preferably 60 %, more preferably 70 %, and even more preferably 80 % of at least one pharmaceutically active agent is prevented from being released substantially immediately (for example, within 30 minutes) from the formulation (see, Figure 8, which is discussed in Example 4). As a result, the formulations of the invention prevent dose dumping in water, 20 %
ethanol, 40 % ethanol, and 60 % ethanol even if the formulations have been broken or crushed.

[0047] In certain embodiments, the formulation of the invention, when crushed and exposed to 900 mL of water in a U.S.P. Type I Apparatus with stirring at 100 rpm for 30 minutes at 37 C, less than about 50 %, less than about 45 %, less than about 40 %, less than about 35 %, less than about 30 %, less than about 25%, or less than about 20 % by weight of at least one pharmaceutically active agent originally present in the formulation before it was crushed broken is released into the water. In certain other embodiments, when the formulation of the invention is crushed and exposed to 900 mL of an aqueous solution containing 60% (v/v) ethanol in a U.S.P. Type I Apparatus with stirring at 100 rpm for 30 minutes at 37 C, less than about 50 %, less than about 45 %, less than about 40 %, less than about 35 %, less than about 30 %, less than about 25 %, or less than about 20 .% by weight of at least one pharmaceutically active agent originally present in the formulation before it was broken is released into the aqueous solution.
[0048] Each of the components of the formulation of the invention are discussed in the following sections.
A. Considerations for the Core [0049] The core comprises a superabsorbent material, which constitutes an important feature of the invention. The term "superabsorbent material," as used herein is understood to mean any material that absorbs solvent, for example, 1 gram of material absorbs at least 30 mL, more preferably 50 mL of solvent, which, upon absorption of the solvent, swells to produce a hydrated gel (hydrogel). In general, useful superabsorbent materials, when exposed to an aqueous medium (for example, water) absorb in excess of 10-15 times, such as at least greater than 30 times, more preferably. 50 times, of water based on its own weight.. In certain embodiments, the superabsorbent material is a polymer.
[0050] Superabsorbent materials can be manufactured from polysaccharide derivatives or cross-linked polyelectrolytes. Polysaccharide superabsorbents include, but are not limited to, a starch graft copolymer, a crosslinked carboxymethylcellulose derivative, a cross-linked hydroxypropyl distarch phosphate, a hydrolyzed starch-acrylonitrile graft copolymer and a neutralized starch-acrylic acid graft copolymer. Cross-linked polyelectrolytes can contain functional groups such as carboxyl, sulfonate, sulphate, sulfite, phosphate, amine, irnine, amide, quaternary ammonium or a mixture thereof. Examples of polyelectolyte polymers include, but are not limited to, salts or partial salts of polyacrylic acid, polyacrylamido rnethylpropane sulfonic acid, polyvinyl acetic acid, polyvinyl phosphonic acid, polyvinyl sulfonic acid, an isobutylene-maleic anhydride copolymer, carboxymethyl cellulose, aiginic acid, carrageenan, polyaspartic acid, polyglutamic acid, polyvinyl amine, polydiallyl dimethyl ammonium hydroxide, polyacrylarnidopropyl trimethyl ammonium hydroxide, polyarnino propanol vinyl ether, polyallylamine, chitosan, polylysine, polyglutamine and copolymers or mixtures thereof 100511 Exemplary superabsorbent materials can include a polymer selected from the group consisting of polycarbophil, polycarbophilic calcium, polymethacrylic acid, polyacrylic acid, and mixtures thereof Polycarbophil is a superabsorbent polymer is capable of absorbing and retaining large quantities of water. Polycarbophil is a high molecular weight acrylic acid polymer cross-linked with divinyl glycol, and is sold under the tradename, NOVEON AA-1, by Lubrizol Corporation OH, USA. It is understood that 1 gram of polycarbophil can absorb about 62 grams of water. Polycarbophil is stable and does not undergo hydrolysis or oxidation under normal conditions. Calcium salts of polycarbophil (polycarbophilic calcium) can be used and are available commercially under the tradename NOVEON CA-1 or CA-2 from Lubrizol Corporation OH, USA. Other exemplary superabsorbent materials include, for example, Carbopol 71G, Carbopol 971P, Carbopol 974 available from Lubrizol Corporation, 0I-I, USA.
[00521 The superabsorbent material provides two functions. First, when the formulation containing the superabsorbent material (for example, polycarbophil) is crushed and combined with solvent (for example, water) for parenteral injection, the superabsorbent material rapidly absorbs the water, swells and forms a hard gel thus preventing injection. In addition, depending upon the amount of solvent added, all of the solvent may be absorbed leaving no residual solvent that can be administered. Second, if the formulation is crushed and snorted into a nostril the superabsorbent material absorbs the liquid in the nostril causing the superabsorbent material to swell. Not only does the swelling cause discomfort but also prevents the drug disposed within the formulation from being rapidly released (for example, within less than 30 minutes).
[0053] In general, the proportion of the superabsorbent material in the core varies from about
10 % (w/w) to about 70 % (w/w) of the core, more preferably from about 30 %
(w/w) to about 50 % (w/w) of the core. Furthermore, the superabsorbent material in the core varies from about 2 % (w/w) to about 20 % (w/w) of the final intact formulation, more preferably from about 4 %

to about 14 % of the final intact formulation, more preferably from about 6 %
to about 12 % of the final intact formulation.
[0054] In addition, relative to the intact formulation, the volume of the core constitutes from about 5 c)./0 to about 40 % of the intact formulation, from about 10 % to about 30 % of the intact formulation, or from about 15 % to about 20 % of the intact formulation. In certain embodiments, the volume of the core constitutes at least 30 %, at least 20 %, or at least 15% of the final volume of the resulting intact formulation.
[0055] In addition to the superab.sorbent material, the core can comprise other excipients and manufacturing aids including, for example, one or more of granulation aids (for example, xanthan gum, polyethylene oxide, polyvinylpyrollidone, cellulose and sucrose derivatives, and mixtures thereof), a lubricant (for example, magnesium stearyl fumarate, magnesium stearate, and stearic acid), a glidant (for example, colloidal silicon dioxide and talc), a dye (for example, iron oxide), and a filler (for example, microcrystalline starch).
[0056] In addition, the core can comprise controlled release microparticles containing a pharmaceutically active agent of interest. Compositions of exemplary controlled release microparticles and methods for their manufacture are described in Section C
below.
[0057] In certain embodiments, the core is monolithic, and optionally comprises microparticles disposed therein. It is understood, however, that under certain circumstances the core can comprise a plurality of different release matrices, which can be, for example, in the form of a bilayer or a multilayer that contains two, three or more layers. One of the layers can act can as an immediate release layer and another layer can act as a controlled release layer. Alternatively, at least two of the layers can have controlled release properties. In one embodiment, one layer can release one pharmaceutically active agent and another layer can release a different pharmaceutically active agent, which can be released at the same or at different rates. In another embodiment, one layer can release one pharmaceutically active agent at one rate and another layer can release the same pharmaceutically active agent at a different rate (i.e., faster or slower than the first layer).

B. Considerations for the Coat [0058] The coat, when present, performs an important function in the operation of the formulation of the invention. The coat provides a hard outer shell that (i) resists damage by crushing or chewing, (ii) resists the release of drug as the pH of the extraction media varies (for example, when the formulations are combined with conventional carbonated beverages), (iii) resists the release of drug in the presence of alcohol in the extraction media even at levels that exceed the alcohol content of alcoholic beverages, and (iv) permits permeation by solvent to permit the release of drug disposed within microparticles located in the core and/or the coat.
Under normal use, the coat still provides a rigid net-like structure that encapsulates the core and prevents the superabsorbent material in the core from swelling.
[0059] In certain embodiments, the coat comprises a controlled release agent.
Alternatively, or in addition, the coat is a controlled release coating. Exemplary controlled release agents and coatings can be selected from the group consisting of acetate suecinate, a polyvinyl derivative (for example, polyvinyl alcohol, polyvinyl acetate, polyvinyl acetate phthalate, a copolymer of vinyl acetate and vinyl pyrrolidone, a copolymer of vinyl acetate and crotonic acid, polyvinylpyrollidone), polyethylene oxide, polyacrylic acid, polysaccharides (for example, modified starch, cross-linked high amylose starch, hydroxypropyl starch, hydroxypropyl methylcellulose phthalate, cellulose and cellulose derivatives (for example, microcrystalline cellulose, carboxymethylethyl cellulose, cellulose acetate, methylcellulose, ethylcellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, cellulose phthalate, cellulose acetate, cellulose acetate phthalate, cellulose acetate propionate, cellulose-acetate succinate, cellulose acetate butyrate, cellulose-acetate trimellitate)), poloxamer, povidone, alginic acid, sodium alginate, polyethylene glycol, polyethylene glycol alginate, gums (for example, xanthan gum), polymethacrylates (including, for example, a copolymer of methacrylic acid and methyl-methacrylate, and a copolymer of methacrylic acid and ethyl acrylate), a copolymer of methacrylie acid and ethyl acrylate, a copolymer of polymethyl vinyl ether and malonic acid anhydride, a copolymer of polymethyl vinyl ether and malonic acid or the ethyl-, isopropyl-, n-butylesters thereof, zein, and mixtures of the foregoing.
[0060] Further examples of controlled release film-coating polymers include, but are not limited to, methylcellulose, ethyleellulose (for example, Aquacoat" type from FMC
Corp.), methylhydroxyethylcellulose, methylhydroxypropylcellulose (for example, Pharmacoat type from Shin Etsu Corp.), ethylhydroxyethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose or methylcarboxymethylcellulose, acrylic polymers, polyvinylacetates, polyvinyl chlorides, polymethylmetacrylates or a teipolymer of vinylchloride, vinylalcohol and vinylacetate, hydroxypropylmethylcellulose phthalate (for example, HP type from Shin Etsu), hydroxypropylmethylcellulose acetate succinate (for example, Aqoat from Shin Etsu), cellulose acetate phthalate (for example, Aquacoat CPD from FMC Corp. or C-A-P NF from Eastman Chemical), polyvinyl acetate phthalate (for example, Sureteric from Colorcon), carboxymethylethylcellulose, and co-polymerized methacrylic acid/methacrylic acid methyl esters (for example, Eudragit from Degussa/Evonik Industries or Kollicoat from BASF or Aciyl-Eze from Colorcon or Eastacryl from Eastman Chemical) [0061] In one embodiment, ICollidon SR (a powder consisting of polyvinyl acetate (8 parts, w/w) and polyvinyl pyrrolidone (2 parts, w/w)) is used in combination with xanthan gum.
kollidon SR is available from BASF, ON, Canada. In another embodiment, the coat can be, for example, Eudragit L3OD 55, available from Degussa/Evonik Industries, NJ, USA.

Furthermore, it is understood that, depending upon the release kinetics desired, the same controlled release agents and coatings can be disposed within or can coat the microparticles described below in Section C.
[00621 In addition, the coat can comprise one or more of a viscosity increasing agent (for example, xanthan gum, polyethylene oxide, polyvinylpyrollidone, cellulose and sucrose derivatives), a lubricant (for example, sodium stearyl fumarate, magnesium stearate and stearic acid), a glidant (for example, colloidal silicon dioxide and talc), and a dye (for example, iron oxide).
[0063] In some embodiments, the coat may comprise a plasticizer.
Examples of plasticizers include, but are not limited to, cetanol, triacetin, citric acid esters, phthalic acid esters, dibutyl succinate, acetylated monoglyceride, acetyltributyl, acetyltributyl citrate, acetyltriethyl citrate, benzyl benzoate, calcium stearate, castor oil, cetanol, chlorebutanol, colloidal silica dioxide, dibutyl phthalate, dibutyl sebacate, diethyl oxalate, diethyl malate, diethyl maleate, diethyl malonate, diethyl fumarate, diethyl phthalate, diethyl sebacate, diethyl succinate, dimethylphthalate, dioctyl phthalate, glycerin, glyceroltributyrate, glyceroltriacetate, glycery1 behanate, glyceryl monostearate, hydrogenated vegetable oil, lecithin, leucine, magnesium silicate, magnesium stearate, polyethylene glycol, propylene, glycol, polysorbate, silicone, stearic acid, talc, titanium dioxide, triacetin, tributyl citrate, triethyl citrate, zinc stearate, PEG (polyethylene glycol), and the like.
[00641 In one embodiment, the coat contains Kollidon SR and xanthan gum as release controlling agents, colloidal silicon dioxide as a glidant, and sodium stearyl furnarate as a lubricant. Incorporation of Kollidoe SR and xanthan gum into the coat helps provide a controlled-release of the pharmaceutically active agent (for example, tramadol HO), and significantly increases the mechanical strength of the resulting formulations making them harder to crush.
[0065] In addition, the coat can comprise controlled release microparticles containing a pharmaceutically active agent of interest. Compositions of exemplary controlled release microparticles and methods for their manufacture are described in the following section.
C. Considerations for the Controlled Release Micropartieles [00661 As shown in Figure 1, the formulations of the invention comprises controlled release microparticles disposed within the coat (Figures 1A and 1D), the core (Figures 1B and 1E), or within both the core and the coat (Figures 1C and IF). The controlled release microparticles contain pharmaceutically active agent and facilitate the controlled release of the pharmaceutically active agent disposed therein. Depending upon the configuration chosen, the formulations can release the pharmaceutically active agent over a prolonged period of time, for example, at least 6 hours, at least 8 hours, at least 12 hours, at least 18 hours, Or at least 24 hours.
[0067] Although the controlled release particles may take a variety of forms, they have a number of features in common, which include (i) they have controlled release properties and (ii) they are of a size that makes them hard to crush even when the formulations are crushed with a conventional pill crusher. The microparticles may have a core and a coat, where either or both provide controlled release properties.
[0068] The core of the microparticles can comprise the pharmaceutically active agent and a variety of' excipients, which include, for example, one or more of, a spheronizing agent, a plasticizer, and a controlled release agent. Exemplary spheronizing agents include, for example, microcrystalline cellulose, ethyl cellulose, low substituted hydroxypropylcellulose and dicalciiun phosphate dihydate. Microcrystalline cellulose is preferred and is available commercially under the tradenarne Avicel PH101 from FMC BioPolymer, DE, USA. Microcrystalline cellulose forms a plastic and cohesive mass upon wetting, which is desirable for the successful production of spherical granules. Microcrystalline cellulose is considered to aid the spheronization process by absorbing water like a molecular sponge and helps in the binding and lubrication of the moistened powder mass during extrusion. During the spheronization process, moisture trapped in the raicrocrystalline cellulose microfibrils adds plasticity to the extrudate and helps convert short round extudates obtained by extrusion into spherical pellets. Different grades of rnicrocrystalline cellulose are commercially available, and a preferred grade suitable for extrusion-spheronization is Avicel PH 101, because of its small particle size, low packing density and high water retentive capacity.
[0069] In addition, the core of the microparticles can contain a plasticizer.
Exemplary plasticizers include, for example, Plasacryl available from IMTech, PA, USA, and triethyl citrate available from Modlex, NC, USA.
[00701 In addition, the core of the microparticles optionally can contain a controlled release agent that controls the release of the pharmaceutically active agent Exemplary controlled release agents can be selected from the group consisting of starch, starch derivatives, cellulose derivatives, xanthan gum, polyethylene glycol, polyvinyl acetate, polyvinyl alcohol, and mixtures thereof. In a preferred embodiment, the controlled release excipient includes a starch derivative that is a cross-linked high amylose starch, which provides the controlled release of the pharmaceutically active agent for at least 12 hours, for at least 18 hours, or for at least 24 hours.
The cross-linked high amylose starch can be cross-linked with phosphorus oxychloride and/or can contain hydroxypropyl side chains. In certain embodiments, a suitable controlled release agent is commercially available from Labopharm, Inc., Laval, Canada, under the trademark CONTRAMID . The synthesis of the CONTRAMID" excipient is described in U.S.
Patent No.
6,607,748.
[0071] The core of the microparticles containing a pharmaceutically active agent can be prepared by a variety of methods, including, for example, wet granulation and extrusion-spheronization.

During wet granulation, microparticles are prepared using, for example, a fluid bed rotor granulator. The wet granulation process comprises, for example, (i) wetting the powder to form wet granules; (ii) exposing the wet granules to tumbling or spheronization, and (iii) drying the product of step (ii). Alternatively, the pellets can be produced by extrusion-spheronization, which has the advantage of being highly reproducible, easy to scale up, cost effective, and produces substantially perfect spherical microparticles. Extrusion-spheronization comprises, for example, (i) wetting the powder blend with an aqueous or organic solution generally containing a binder to form a wet homogeneous mass suitable for wet extrusion, (ii) extruding the wet mass to form cylindrical extrudates of uniform shape and size, and (iii) spheronizing the wet extrudates using a spberonizer, where, for example, a fast spinning disc, breaks the extrudates into smaller microparticles and rounds them to form spheres.
[0072] The cores of the microparticles can be coated with a controlled-release coating that is sufficiently flexible to be deformed under compression during tablet formation without undergoing fracture. Suitable controlled release agents are described in the previous section. In one embodiment, the controlled release coating comprises polymethacrylate (e.g., Eudragit RS, available from Degussa/Evonik Industries, NJ, USA). Eudragit RS3OD grade, which is particularly useful is an aqueous dispersion (30% w/vv) of a polymeric mixture of ethyl acrylate, methyl methacrylate, and trimethylarnmonioethyl methacrylate at a ratio of 1:2:0.1 (w/w). The Eudragit RS grade is designed to form water-insoluble film coats for sustained release formulations, The Eudragit RS grade forms a highly flexible film coat with low permeability.
Another useful coating material includes Eudagrit 1.,30D 55, available from Degussa/Evonik Industries, NJ, USA. Another controlled release coating comprises ethyl cellulose sold under the tradename Surelease . Another controlled release coating includes Kollicoat SR, available from BASF Fine Chemicals, In one approach, the core of the microparticles is coated using a fluid bed coater equipped with a bottom spray.
[0073] The resulting particles, depending upon their composition and method of fabrication have an average diameter in the range of from about 1 j.im to about 1000 um. In certain embodiments, the microparticles have an average diameter of from about of 200 pm to about 900 p.m, or from about 300 pm to about 800 pm. In certain embodiments, the resulting microparticles have an average diameter of about 700 p.m In certain other embodiments the microparticles have an average diameter of from about 1 p.m to about 400 pm, from about of 5 um to about 300 pin, or from about 10 pm to about 200 inn. In certain embodiments, the resulting microparticles have an average diameter of about 100 um.
D. Pharmaceutically Active Agents [0074] It is understood that the compositions described herein can be used for the delivery of one or more pharmaceutically active agents. In certain embodiments, the controlled release microparticles can contain one or more pharmaceutically active agents. In addition, it is understood that the formulations of the invention can contain a number of different microparticles, with one population of microparticles containing one pharmaceutically active agent and another population of microparticles containing a second, different pharmaceutically active agent.
[0075] Many pharmaceutically active agents can benefit from being delivered using the formulations described herein. The Controlled Substances Act (GSA), Title II
of the Comprehensive Drug Abuse Prevention and Control Act of 1970, places all substances that are regulated under existing Federal Law into one of five schedules based upon the substance's medicinal value, harmfulness, and potential for abuse or addiction. The formulations of the invention are preferably used to deliver those drugs classified as Schedule II, III, IV and V drugs.
Similarly, although any drug in which there is a benefit in having controlled release of the drug can be incorporated into formulations of the invention, the formulations described herein are particularly useful in the delivery of, for example, CNS and respiratory stimulant agents, analgesics (for example, opioid analgesics), hypnotic agents, anxiolytic agents, and agents with a narrow therapeutic index. For purposes of this invention, pharmaceutically active agents are intended to encompass salts, esters, and the prodrugs of the pharmaceutically active agents.
[0076] Exemplary opioid analgesics include, for example, alfentanil, buprenorphine, butorphanol, carefentanil, codeine, dezocine, diacetylmorphine, dihydrocodeine, dihydromorphine, diprenorplune, etorphine, fentanyl, hydrocodone, hydrornorphone, I3-hydroxy-3 -methylfentanyl, levo a-acetylmethadol, levorphanol, lofentanil, meperidine, methadone, morphine, nalbuphine, oxycodone, oxymorphone, pentazocine, pethidine, propoxyphene, remifentanil, sufentanil, tilidine, tramadol hydrochloride, or a mixture thereof.
[0077] Exemplary hypnotics include, for example, benzodiazepines and non-benzodiazepines.
Exemplary benzodiazepines include, but are not limited to, alprazolam, diazepam, flurazepam, loprazolam mexazolam, nitrazepam, and the like. Exemplary non-benzodiazepines include, but are not limited to, barbiturates (for example, butobarbitone, phenobarbitone,.
or arnylobarbitone) chlonnethiazole, eszopiclone, ramelteon, zaleplon, zopiclone, zolpidem, and the like.
100781 Exemplary anxiolytic agents include, but are not limited to, amphetamine, buspirone, barbiturates, benzodiazepines (for example, alprazolan, bromazepam, brotizolatn, camazepam, chlotdiazepoxide, clobazarn, clonazepam, desalkylflurazepain, diazepam, flunitrazepam, flurazepam, lorazeparn, lometazepam, medazepam, metaclazepam, midazolam, nitrazeparn, nordazepam, oxazepam, pentylenetetrazole, prazepam, temazepam, tetazepam, and triazolain) and the like.
[0079] Exemplary CNS and respiratory stimulatory agents include, but are not limited to xanthines (for example, caffeine and theophylline), amphetamines (for example, amphetamine, benzphetamine hydrochloride, dextroaniphetarnine, dextroamphetamine sulfate, levamphetatnine, levamphetamine hydrochloride, methamphetamine, and methamphetamine hydrochloride), and miscellaneous stimulants such as methylphenidate, rnethylphenidate hydrochloride, modafinil, pemoline, sihutramine, and sibutramine hydrochloride.
100801 Pharmaceutically active agents with a narrow therapeutic index include, for example, amiodarone, anaphotericin, cabamazepine, clozapine, digoxin, disopyramide, lithium carbonate, mmoxidil, phenytoin, primidone, procainamide, quinidine, theophylline, valproic acid, and warfarin.
[00811 It will be appreciated that the amount of the pharmaceutically active agent present in the abuse-resistant formulation depends upon the therapeutic dose required in conventional tablets In generally, each pharmaceutically active agent is present in an amount ranging from about 0.5 mg to about 900 mg by weight, from about 1 mg to about 700 mg by weight, from about 1 mg to about 600 mg by weight, from about 1 mg to about 500 mg, from about lmg to about 400mg, from about 1 mg to about 300 mg, from about 1 mg to about 200 mg, and from about 10 mg to about 200 mg. It is understood, however, that the actual dosage will depend upon the particular pharmaceutically active ingredient and its proposed use.
[0082] The invention also provides a solid dosage form for the controlled release of a pharmaceutically active agent disposed therein. The solid dosage form comprises an admixture of a superabsorbent material (for example, polycarbophil) and a plurality of controlled release microparticles having a pharmaceutically active agent disposed therein. When the solid dosage form is exposed intact to an aqueous environment, the pharmaceutically active agent is released from the solid dosage form over a prolonged period of time. However, when the solid dosage form is crushed to expose the interior of the core and exposed to an aqueous environment, the superabsorbent material swells to create a hard gel that traps the microparticles, and the microparticles provide controlled release of the pharmaceutically active agent The solid dosage form can be coated or uncoated. Accordingly, it is understood that the features and components of the coated formulations described hereinabove are also applicable to the solid dosage form.
[00831 It is understood that the intact compositions described herein can be produced using techniques known to those in a formulary arts. An exemplary protocol for producing controlled release tablets is described in Example 1. It is understood, however, that other approaches can be used to make formulations of the invention. The formulations of the invention preferably have a hardness in the range of from about 100 N to about 500 N, or from about 150 N to about 400N, or from about 200 N to about 400N, or from about 300 N to about 400 N, with a target hardness of at least 200 N. Furthermore, the formulations of the invention may take the form of capsules, caplets, tablets, or pills.
[0084] The formulations of the invention can be used to administer a pharmaceutically active agent to a mammal, for example, a human, in need of the pharmaceutically active agent (for example, an opioid analgesic for pain management). It is understood that the exact dosage will vary depending on the symptoms, age, body weight, severity of the disease to be treated and can be optimized through routine experimentation known to those of skill in the art.
EXAMPLES
[0085] Practice of the invention will be more fully understood from the foregoing examples, which are presented herein for illustrative purposes only, and should not be construed as limiting the invention in any way.

EXAMPLE 1 - Preparation of Exemplary Tramadol Containing Controlled Release Formulation [0086] This Example describes an exemplary misuse preventative tablet and how it can be made.
The formulation contains tramadol, an opioid drug used for the treatment of moderate to moderately severe pain, which is capable of being abused and for which over exposure via misuse can lead to harmful side effects. The misuse preventative tablet described in this Example contains 100 mg of trarnadol HCI which, as can be seen from Example 2, is released from the intact tablets over 24 hours The formulation of the complete tablet is set forth in Table 1, and the manufacture of each of the components for the formulation appear in the following sections of this Example.

Component Mg / Tablet ' Core blend Coat blend Tramadol HC1 25.0 75.0 Avicel PH 101 30.6 30.0 Contramid 0.7 2.1 Polycarbophil (Noveon AA-1) 62.9 Xanthan gum 20.6 241.6 Kollidon SR 120.5 Eudragit RS 30D 5.7 17.1 Triethyl citrate 0.6 1.7 Plasacql0.9 2.6 . _ .
Colloidal silicon dioxide 0.75 2.5 Sodium stearyl fumarate 1.5 5.0 FD&C Blue #1 Aluminium lake 11-13 0.08 Opadry white 0.67 21.3 ¨
[0087] The formulation of Table I was prepared by a multi-step process, which is outlined below in subsections A-D.
A. Manufacture of Trarnadol Containing Controlled Release Microparticles [0088] The formulation of uncoated microparticles is set forth in Table 2, and the uncoated microparticles were produced as follows. The various components were mixed in a mixer for 3 minutes under low shear conditions. The dry blend then was wetted under agitation in the same mixer by gradually adding water until a homogeneous wet mass suitable for extrusion was produced. The wet mass then was extruded at a constant speed (45 rpm) using a Laboratory Multigranulator extruder model MG-55 from LCI, Inc., NC, USA equipped with a dome die having a 0.6 mm diameter hole and a fixed extrusion gap. The extrudes then were spheronized at a constant speed (1800 rpm) using a Maturnerzier Model ()J.-230T from LCI, Inc., NC, USA.
The wet microparticles were dried at 45 C in a fluid bed until a moisture content of about 2 %
was achieved.

COntlionent .: . . Wejglat in Uncoatcd. Weight .(g): in ! : = !Mtcroparticles = = hatch ; = 1 Tramadol HCI 70.0 2,800.0 Avicel PH-101 28.0 1,120.0 Contramid 2.0 80.0 Water 600.0 I Total 100 4000.0 [0089] The resulting microparticles then were coated with a controlled release coating and an Opadry II White containing film as described in Table 3. The microparticles were coated in a fluid bed coater. The microparticles were film coated to a weight gain of between 7% and 15%
using an aqueous solution of Eudragid RS30C containing Plasacryl and niethyl citrate (TEC).
Afterwards, a curing solution containing Opadry II White was added to provide a film around the Eudragit containing coat to reduce the likelihood of the microparticles sticking together.

_gio = Siihiiiint PirMiCrOtiai;ticics = .
Components ________________ Dry Quantity 'weighed (0'..
Uncoated pellets 1000.0 Eudragit RS 30D ; 160.0 533.3 TEC ! 24.0 24_0 _Plasacryl 16.0 80.0 Opadry II White 118.0 I 18-0 [0090] The resulting controlled released microparticles had a mean diameter of about 700 Lim as measured by an optical microscope.

B. Manufacture of Core Composition 100911 In addition to the controlled release micropartieles, the core contained polycarbophil as well as several other components. The remaining excipients for the core are set forth in Table 4, and were mixed and subjected dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi.

CoMponents :% by Weiglit :in -Quantity. Weiglied :1. H Core GrauubtiOn .(g) Polycarbophil 59.80 1794.0 Avicel PH-101 19.85 595.5 Xanthan Gum 19.85 595.5 Colloidal silicon dioxide 0.25 7.5 Sodium stearyl fumarate 0.25 7.5 Total 100.0 3000.0 [0092] The tramadol containing tnicroparticles then were mixed with the remaining granulated core excipients to produce the formulation of the core, which is set forth in Table 5.

= = . FOrnyolation COmpo ition . .
Core Blend ,õ ! = . ; oks of ' Granulation =, :
Mg/Tablet . . Core:. gibatch Tramadol Containing Microparticles 43.5 29.00 464.0 Polycarbophil 62.58 Xanthan gum 20.72 Granulated Avicel PH 101 20.82 Excipients Colloidal silicon 104.7 0.26 69.79 11166 dioxide Sodium stearyl fumarate _ Colloidal silicon dioxide 0.5 _ 0.33 5.3 Sodium stearyl fiimarate 1.2 0.83 13.3 PD&C Blue #1 Aluminium Lake 11-13 0.1 0.05 0.8 Total. 150.0 100.00 1600 C. Manufacture of Coat Composition [0093] In addition to the controlled release microparticles, the coat contained Ko1lidon6 SR and xanthan gum as well as several other components The remaining excipients for the coat are set forth in Table 6, and were mixed and subjected to dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi.

Components , % b*--Weight in Coat Quantity Weighed (g) Granulation :. = .
Crospovidone (Kollidoe SR) 33.17 995.1 Xanthan gum 66.33 1989.9 Colloidal silicon dioxide 025 7.5 Sodium stearyi fumarate 0.25 7.5 Total 100.00 3000.0 f00941 The tramadol containing microparticles then were mixed with the remaining granulated coat excipients to produce the formulation of the coat, which is set forth in Table 7.

= . Formulation Com osition = :
Coat blend g Granulatioa 1 / 'of . ; = /tabletgmateli Ceat Tramadol Containing 130.5 26.10 1409.4 Micropartieles Kollidon SR 120.46 Xanthan gum 240.88 Granulated Colloidal silicon Excipients 363.8 tiXJ1 72.75 3928.3 dioxide Sodium stearyl 0.91 fumarate Colloidal silicon dioxide 1.6 0.32 17.3 Sodium stearyl fumarate 4.2 0.83 44.9 Total 500.0 100.00 5400.0 D. Tablet Manufacture (00951 Dry-coated tablets then were prepared using a Dry-Cota 16-Station tablet press from Manesty, UK. The core formulation was added to a first hopper in the tablet press and compressed into a core tablet. The coat formulation then was added to a second hoppei in the tablet press and the core and the coat were compressed together to form the thy coated tablet.
The resulting dry coated tablets then were film coated with a solution of Opadry II using a fully perforated pan coating machine (O'Hara, Mississauga, ON, CA) The formulation of film coated tablets is set forth in Table 8.

Components Quantity weighed (g) Dry Coated Tablets 2000.0 Opadry II White solution (20%) 60.0 [0096] The resulting tablets had a hardness in the range of from about 300 N
to about 400 N
with a target hardness of about 350 N.
EXAMPLE 2 - Release Properties of Intact Tablets [0097] The release kinetics of the intact tablets produced in Example 1 were studied in this Example. In addition, the release kinetics were studied when alcohol was included in the extraction media and also when the pH of the extraction media was varied.
[0098] Initially, tramadol release was measured using the rotating basket method (L.T.S.P. Type I
Apparatus) as described in ti S.P. 30 at 100 rounds per minute, at 37+0.5 C, in 900 nal, of potassium phosphate monobasic pH 6.8 solution (buffer stage) during 24 hours.
The results from three experiments are summarized in Figure 2. As can be seen from Figure 2, the tablets produced in Example 1 release tramadol over a 24 hour period with kinetics summarized in Table 9.

Time (hours) I04 Tramadol Standard Release Deviation 0.5 4 0.4 1.0 8 0.7 2.0 17 1.6 4.0 31 2.6 7.0 46 2.9 9.0 55 2.S
12.0 64 2.3 16.0 73 1.8 Time (hours) % Tramadol Standard 'Release I 'Deviation 20.0 80 1.2 24.0 85 1.0 [0099] From the release kinetics presented in Figure 2 and summarized in Table 9, the tablets produced in Example 1, under the conditions tested, released Tramadol over 24 hours with quasi-zero order release kinetics.
[00100] In addition, the effect of alcohol on the release kinetics were studied under the same conditions as before except the extraction solvent was vaned to include water, 20 % ethanol in water, 40 % ethanol in water, 60 % ethanol in water, 80 % ethanol in water and 100 % ethanol.
The results are set forth in Figure 3, which shows that over 6 hours, less than about 30 % of the trarnadol was released when the extraction solvent contained up to 60 %
ethanol, The tablets performed similarly when exposed to water, 20 % ethanol, 40 % ethanol and 60 %
ethanol.
However, about 50 % of the tramadol was released over 6 hours when the tablets were exposed to extraction solvents containing SO % and 100 % ethanol.
[001011 The results set forth in Figure 3 demonstrate that the controlled release properties of the tablets produced in Example 1 was maintained in extraction solvents containing 100 % water or 100 % ethano1_ In some cases, for example, in the presence of 20 % ethanol, the release rate was even slower than in water. Furthermore, under the conditions tested, less than 20 % of the Tramadol was released from the intact tablets in 30 minutes when placed in water, 20 % ethanol, 40 % ethanol, 60% ethanol, SO % ethanol, or 100 % ethanol. Accordingly, it appears that the formulations of the invention are compatible with conventional alcoholic beverages.
[001021 In addition, the effect of pH on the release kinetics were studied under the same conditions as before except the extraction solvent was varied to include water, phosphate buffer at pH 6.8, phosphate buffer at pH 5.0, phosphate buffer at pH 3.0, and acidified water at pH 1.2.
The results are set forth in Figure 4, which shows that the controlled release properties of the tablets produced in Example 1 were maintained as pH was reduced to 1.2. It appears, however, that the rate of release increased as pH decreased from 6.8 to 1.2.
Accordingly, it appears that the formulations of the invention are compatible with various common beverages (for example, carbonated drinks) that have a pH of about 3.5.

EXAMPLE 3 - Release Properties of Bisected Tablets [00103] This Example demonstrates that, under the conditions tested, the tablets produced in Example I can be bisected without destroying the controlled release properties of the tablet. In other words, dose dumping did not occur when the tablets were broken in half.
[00104] Briefly, tablets produced in Example 1 were bisected in half. The release kinetics of the intact tablets and the halves of the bisected tablets were measured in a U.S.P. Type I Apparatus.
The results were normalized for the bisected tablets and are summarized in Figure 5. The kinetics of tramadol release from an intact tablet and a bisected tablet in a Type I Apparatus are Summarized in Table 10 and Table 11, respectively.

- - , Time (hour) = %=Tramariel neiebise 'Standard ;
0.5 4 0.4 1.0 9 0.9 2.0 20 1.2 4.0 38 1.6 7.0 55 2.7 9.0 64 1.5 12.0 72 4.0 16.0 79 4.4 20;0 84 4.8 -24.0 90 6.6 (hours) 10Traniadol Release : Standard . . = Deviation 0.5 9 1.5 1.0 16 2.4 2.0 29 3.7 4.0 48 5.6 7.0 68 7.4 9.0 ' 76 9.5 12.0 88 7.4 16.0 94 7.8 20.0 j98 8.2 24.01,100 8.5 100105] Figure 5 demonstrates that both the intact tablet and the bisected tablet maintain their controlled release properties and release tramadol over 20-24 hours. The release profile for the bisected tablets was similar to that of the intact tablets, however, it appeared that the bisected tablets released the tramadol slightly faster than the intact tablets. For example, at the 12 hour time point, the bisected tablet released 80-90 % of the starting amount of tramadol whereas the intact tablets released 65-75 % of the tramadol.
EXAMPLE 4 - Release Properties of Crushed Tablets [00106] This Example describes the performance of the tablets made in Example 1 after crushing with a conventional pill crusher. In particular, the performance of the crushed tablets was measured after being exposed to a number of extraction solvents under different conditions.
[001.071 Initially, the tablets produced in Example 1 were crushed with a pill crusher and combined in a glass vial with 2 mL of water (a volume typical for intravenous drug abuse and greater than the volume typically available if the crushed tablet is mixed with food). The experiment was performed using 4 different lots of tablets. Once the crushed tablet was combined with 2 mL of water, a hard gel was created within 20-30 seconds at the bottom of each leaving no available liquid that could be drawn into a syringe. As shown hi Figure 6, the vials could be inverted and the hard gels remained at the bottom of each vial. In Figure 6, the first vial contained 2 mL of water and vials 2-5 (inverted) contained crushed tablets from four separate lots (denoted Lots 1-4) each combined with 2 mL of water. In each case, the gel produced was rigid enough to remain at the bottom of the vial even when inverted.
[001081 In addition, the ability to extract tramadol from the tablets produced in Example 1 was tested under different conditions after each tablet had been crushed with a pill crusher. Briefly, the crashed tablet was combined with 10 mL of extraction media (water, acid, base, or alcohol containing solvent) in a vial. The solution was heated to the specified temperature (room temperature (RT), 50 C, 75 C, or 100 C) and agitated mechanically for 15 minutes using a wrist action Burrell agitator. It was found, however, than no residual supernatant was produced.
Figure 7 shows seven inverted vials, each containing a hard gel produced after a tablet prepared in Example I had been crushed in a pill crusher and exposed to 10 ml, of extraction media and incubated under various conditions, which included (1) water at room temperature for 15 minutes (Vial 1, Figure 7), (2) water at 50 C for 15 minutes (Vial 2, Figure 7), (3) water at 75 C for 15 minutes (Vial 3, Figure '7), (4) water at 100 C for 15 minutes (Vial 4, Figure 7), (5) acidic media (acidified water) at room temperature for 15 minutes (Vial 5, Figure 7), (6) basic media (sodium hydroxide pH 10) at room temperature for 15 minutes (Vial 6, Figure 7), and (7) 40%
ethanol at room temperature for 15 minutes (Vial 7, Figure 7) As can be seen in Figure 7, all of the conditions tested resulted in formation of hard gels that remained at the bottom of each vial upon inversion. There was no residual supernatant produced by this process and so it was not possible to measure how much tramadol, if any, had been released from the formulation.
[00109] In another experiment, the release of tramadol was measured from tablets produced according to Example 1 after they had been crushed and exposed to solutions containing different concentrations of ethanol (20 %, 40 % and 60 A ethanol). Briefly, the tablets were crushed and the amount of drug release into 900 mL of extraction media in a U.S.P. Type Apparatus with stirring at 100 rpm at 37 C over 30 minutes. The results are summarized in the bar chart appearing in Figure 8. In addition, the extraction of tramadol from commercially available Ultram ER was measured once the Ultram ER had been crushed and exposed to water under the same conditions as those used for the tablets produced in Example 1.
[00110] The results summarized in Figure 8 show what there is no dose dumping of tramadol from the tablets of the invention when exposed to 900 mL, of water, 20 %
ethanol, 40 % ethanol or 60 % ethanol. Under the conditions tested, less than 20 % of the tramadol was released after 30 minutes. In contrast, when commercially available Ultram ER was tested under the same conditions using water as the extraction media, approximately 80 % of the tramadol was released.
[00111] In another experiment, the release of tramadol was measured from tablets produced according to Example 1 after they had been crushed and exposed to extraction media having different pH values, which included water, phosphate buffer at pH 6.8, phosphate buffer at pH 5, phosphate buffer at pH 3, and acidified water at pH 1.2. The tablets were crushed and the amount of drug release into 900 mL of extraction media in a U.S.?. Type I
Apparatus with stilling at 100 rpm at 37'C over 30 minutes. The results are summarized in the bar chart appearing in Figure 9. In addition, the extraction of tramadol from commercially available Ultram ER was measured once the Ultram ER had been crushed and exposed to water under the same conditions as those used for the tablets produced in Example 1.

[00112] The results summarized in Figure 9 show that, under the conditions tested, there was no dose dumping of tramadol when incubated in 900 mL of extraction media (including water, phosphate buffer at pH 6.8, pH 5.0 or pH 3.0, and acidified water at pH 1.2).
It is noted, however, that, under the conditions tested, as pH decreased the amount of released tramadol increased. For example, in water, less than 20 % of the tramadol was released.
In contrast, when commercially available Ultram ER was tested under the same conditions using water as the extraction media, approximately 100 % of the tramadol was released. In phosphate buffer at pH
6.8, 5, and 3, approximately 30-35 % of the tramadol was released from the tablets of the invention, and in acidified water at pH 1.2 approximately 65 % of the tramadol was released.
EXAMPLE 5 ¨ Pharmaeokinetie Properties of Tramadol Tablets [00113] The pharmacokinetic properties of the 100 mg tablets prepared in Example 1 were assessed in a single dose, randomized, crossover study in 18 healthy adults under both fasting conditions and fed conditions After administration, plasma samples were harvested periodically, and the concentration of tramadol present in the plasma was measured via liquid chromatography-tandem mass spectrometry.
[00114] The results were plotted in Figure 10, where the mean plasma concentrations of tramadol present in the plasma under fasting conditions is shown in Figure 10A
and the mean plasma concentrations of tramadol present in the plasma under fed conditions is shown in Figure 10B. The median Tm. (hours) was 6.0 hours for both the fed and fasted conditions, The Cõ,õõ
(ng/mL) was 120 32 nem', and 154 41 ng/mL under fasted and fed conditions, respectively.
The T)/2 (hours) was 8.4 2.9 hours and 6.8 2.1 hours following fasted and fed administration, respectively. The AUCo.f (ngh/rnL) was 2556 1026 and 2746 1057 for the fasted and fed states, respectively, and the AUC0, (ngh/mL) was 2703 1109 and 2829 1119 for the fasted and fed states, respectively.
EXAMPLE 6¨ Exemplary Oxycodotie Tablet [00115] This Example describes the manufacture and testing of 40 mg oxycodone HC1 tablet (BID) having a core and a controlled release coating. The coat comprises microparticles that provide controlled release properties and reduce misuse of the oxycodone disposed within the micropartieles.

(00116) The microparticles were produced by extrusion spheronization, which produces the microparticles, and then were coated by fluidized bed coating. The resulting coated microparticles were blended with the coat matrix excipients and then compressed around a pre-formed polycarbophyl core.
[00117] The composition of the oxycodone containing microparticles are set forth in Table 12 Table 12 fiPiripaZajalpi ViOtteltailtig Eµjf;i10.5.-:-.INV:a54411:161151116C.1,4t4iiiP.100411 Avicel PH 101 72.000 -Contramid 2.297 Eudragit RS 30D 9.136 .
Triethyl citrate 1.365 ._..
Plasacryl 0.906 , Oxycodone HC1 , 40.000 [001181 The resulting microparticles then were coated in a fluid bed coater equipped with a bottom spray. The microparticles were film coated to a weight gain of 7% to 15% using an aqueous solution of Eudragid RS30C containing Plasacryl and triethyl citrate (TEC).
Afterwards, a curing solution containing Opaciry II White was added to provide a film around the Eudragit containing coat to reduce the likelihood of the microparticles sticking together, [00119] The composition of the core and the coat is set forth in Table 13.
Table 13 1:14 q ___________________________________________________________ --9zo7 _.= 1"51P-::51-Z¨i;41Nirgr-7-E-2 :i.
7 ,4171Z4M-217.%4112130,M
:.=j-j:=r, 7---;i,-:Z: ________________ 1' -- -=.-E.??!!.10:irA=.-;- ..
EiliTS:2111i';':riNMW-44A522initiWleatti :giiT- if?: gr, i'l-Fila MOM
0 codone Ha . ovided as micro .articles __ 125.704 125.704 Avicel PH 101 13.749 ..... 13.749 Contramid IMIIIIII 2.297 2.297 Polycarbo chit (Noveon AA-1 41.420 --- 41.420 Xanthan . um 13.749 14.451 48.200 KollidonZ SR ________________________________ 68.908 68.908 Colloidal silicon dioxide 0.349 1.440 1.789 Sodium ste: 1 fumarate 0.698 2.610 3.308 FD8cC Yellow #6 Aluminium lake 0.035 _,__. . 0.035 Total 70.000 233.113 303.113 [00120) The dry-coated tablets were prepared using a Dry-Cota 16-Station tablet press from Manesty, UK. The core formulation was added to a first hopper in the tablet press and compressed into a core tablet. The coat formulation then was added to a second hopper in the tablet press and the core and the coat were compressed together to form the dry coated tablet.
The resulting dry coated tablets then were film coated with a solution of Opadry II using a fully perforated pan coating machine (O'Hara, Mississauga, ON, CA).
[00121] The in vitro release properties of the resulting tablets were measured in a U.S.P. Type I
Apparatus in phosphate buffer pH 6.8 or water containing 40 % ethanol. The release kinetics were measured on intact tablets (see, Figure 11A) or crushed tablets (see.
Figure 1113), which had been crushed by using a conventional pill crusher. Figure 1113 also shows the release of oxycodone over time from Oxycontin tablets available commercially from Purdue Pharma. In addition, the release kinetics were measured for intact tablets in the presence of phosphate buffer pH 6.8, and for bisected tablets (half tablets) in the presence of phosphate buffer pH 6.8 (see, Figure 12). As shown in Figure 12, the release profiles were substantially the same for the intact tablets and the bisected tablets.
[00122] The intact tablets provided controlled release over 12 hours and the release was not materially affected by the presence of 40 % ethanol. In contrast to the crushed Oxycontin tablets, neither the crushed nor the bisected tablets (half tablets) produced in accordance with the invention released oxycodone by dose dumping, and no dose dumping was seen in the presence of 40 % ethanol.
EXAMPLE 7¨ Exemplary Oxycodone HO/Acetaminophen Tablet [00123] This Example describes the manufacture and testing of a twice a day tablet (BID) containing 20 mg oxycodone HC1 and 650 mg of acetaminophen. The tablet comprises a core surrounded by an enteric, controlled release coating (namely, Eudragit L30D55), where the core is in the form of a bilayer.
The composition of the microparticles is set forth in Table 14.

Oxycodone 11C1 20.0 11.51 Cellulose micromystalline (Avicel PHI 01) 37.3 21.49 Contramid 2.7 1.53 Lactose monohydrate 73.4 42.22 Eudragit NE 300 20.0 11.51 Talc 20.0 11.51 Colloidalsilijon dioxide 0,4 0.23 Total 173.8 100.00 (001241 The microparticles were produced by mixing the components set forth in Table 14 (except for the Eudragit NE 30D and Talc). The resulting mixture was subjected to extrusion and spheronization, and the resulting microparticles were coated with the Eudragit NE 30D and talc in a fluid bed coater equipped with a bottom spray. The core of the tablet was a bilayer. The oxycodone containing microparticles were incorporated in the slow release layer of the bilayer whereas the acetaminophen, as COMPAO which was in free form and not incorporated into microparticles, was present in both the rapid release layer and the slow release layer.
1001251 The composition of the bilayer core is set forth in Table 15.

Ingreilient5 . . Tablet ComPosition First layer (rapid release) (Mg) ICA) COMPAP (which includes acetaminophen) '288.89 89.72 Microcrystalline Cellulose PHI 02 19.77 6.14 Croscaramellose sodium AeDiSol 6.70 2.08 Colloidal silicon dioxide (Cab 0 sit) 1.68 052 Sodium stearyl fumarate (Pruv) 4.83 1.50 FD&C Yellow 116 0.13 0.04 Total 322.00 100.00 Ingredients = Tablet composition Second layer Wow (Mg) I(%) Oxycodone (provided as oxycodone microparticles) 173.79 24.72 compApt (which includes acetaminophen) 43133 61.64 Carbopol 71 G ,42,02 5.98_ Xanthan gum 80 mesh 42.02 5.98 Colloidal silicon dioxide (Cab 0 sil) 2.95 0.42 Sodium stearyl fumarate (Pruv) 8.86 1.26 Total 703.00 100.-00 [00126] The bilayer core was prepared by mixing the components of each layer and then compressing the materials in a PiecolaTM bilayer tablet press (SMI Inc., NJ, USA). The bilayer tablets had a hardness in the range of 190 to 230 Newtons. The resulting bilayer core was then coated with Eudragit 1_,30D 55 by using a fully perforated pan coating machine (O'Hara, Mississauga, ON, CA). The resulting coating contained 82 mg of Eudragit 1_,30D
55, which accounted for 8% of the weight of the tablet, (00127] The in vitro release kinetics of the resulting tablet were measured in a U.S.P. Type III
Apparatus at 20 dprn after incubation in 0.1M hydrochloric acid at pH 1.2 for 1 hour followed by incubation in phosphate buffer pH 6.8 for 11 hours. The results shown in Figure 13 indicate that no oxycodone was released from the tablet for about one hour when the tablet was in 0.1 M
Once the pH was raised after one hour, the oxycodone was released with controlled release kinetics.
EXAMPLE 8¨ Exemplary Once-a-Day 150 mg Tramadol Tablet [00128] This Example describes the manufacture and testing of an exemplary once-a-day 150 mg tramadol HCI tablet, where the tablets have a monolithic core and a controlled release coating. The core comprises a super absorbent polycarbophil and the controlled release coat comprises xanthan gum and Kollidon. Tramadol containing microparticles are disposed within both the core and the coat.
[00129) The composition of the microparticles is set forth in Table 16_ Ingredients . , compoiition Tramadolliel 57.38 MCC Avicel PH 101 24.60 Eudragit RS30D + Plasacryr + Triethyl citrate 16.39 Opadryllwhite L63 Total 100.00 [00130] Uncoated microparticles were produced as follows. Tramadol and Avicel PH 101 were mixed in a mixer for 3 minutes under low shear conditions. The dry blend then was wetted under agitation in the same mixer by gradually adding water until a homogeneous wet mass suitable for extrusion was produced. The wet mass then was extruded at a constant speed (45 rpm) using a Laboratory Multigranulator extruder model MG-55 from LCI, Inc., NC, USA
equipped with a dome die having a 0.6 mm diameter hole and a fixed extrusion gap. The extrudates then were spheronized at a constant speed (1,800 rpm) using a Maturnerzier Model Q.1-230T from LCI, Inc., NC, USA. The wet microparticles were dried at 45 C in a fluid bed until a moisture content of about 2 % was achieved.
[00131] A portion of the resulting microparticles were coated with an aqueous solution containing Eudragit RS 30D using a fluid bed coater. The microparticles were film coated to a weight gain of between 7% and 15%. Afterwards, a curing solution containing Opadry II White was added to provide a film around the Eudragit containing coat to reduce the likelihood of the microparticles sticking together.
[00132] The composition of the core granules is set forth in Table 17.

Ingredients ' : % Cm_pri osition Polycarbophilic acid (Noveon AA-1) 80.00 MCC PH-101 19.50 Colloidal silicon dioxide 0.25 Sodium stearyl fumarate 0.25 Total 100.00 [00133] In addition to the controlled release microparticles, the core contained polycarbopbil as well as several other components. The remaining excipients for the core were mixed and subjected dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi. Then, uncoated microparticles were mixed with the granulated core excipients to produce the core formulation.
[00134] The composition of the coat granules is set forth in Table 18.

Ingredients Composition Kollidon SR 49.75 Xanthan gum 49.75 Colloidal silicon dioxide 0.25 Sodium stearyl fumarate 0.25 Total 100.00 [00135] The remaining excipients for the coat were mixed and subjected to dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi. Then, coated microparticles were mixed with the granulated coat excipients to produce the coat formulation [00136] The composition of the tablet is set forth in Table 19.

= . ' ' Composition .
ingredients Mg / tablet -Core formulation -Tramadol HCI microparticles 36.31 65.36 Core granules 62,44 112.39 Colloidal silicon dioxide 0.50 0.90 Sodium stearyl fmnarate 0.75 1.35 Total 100 180 Coat formulation Tramadol HCl microparticles (film coated) 35.98 196.09 Coat granules 63.02 343.46 Colloidal silicon dioxide 0.25 1 .36 Sodium stearyl fumarate 0.75 4.09 Total 100 545 [00137] Dry-coated tablets then were prepared using a Dry-Cota 16-Station tablet press from Manesty, UK. The core formulation was added to a first hopper in the tablet press and compressed into a core tablet. The coat formulation then was added to a second hopper in the tablet press and the core and the coat were compressed together to faun the dry coated tablet.
The resulting dry coated tablets then were film coated with a solution of Opadry rr using a fully perforated pan coating machine (O'Hara, Mississauga, ON, CA).
[00138] The in vitro release properties of the resulting tablets (both intact and crushed) were measured in a U.S.P. Type I Apparatus in phosphate buffer pH 6.8. Three separate batches were tested The results of in vitro release from the intact tablets is shown in Figure 14A and from crushed tablets is shown in Figure 14B. The tablets were crushed using a pill crusher. The results show that the intact tablets of the invention demonstrated a controlled release of tramadol over 24 hours in phosphate buffer pH 6.8. Moreover, there was no dose dumping of tramadol from the crushed tablets when exposed to the same dissolution conditions.
Under the conditions tested, less than 50 % of the tramadol was released within 60 minutes.
[00139] In addition, the in vitro release properties of the resulting tablets (both intact and crushed) were measured in a U.S .P Type I Apparatus in water or water containing 20 % ethanol, 40 % ethanol and 60 % ethanol. The same three batches were tested. The results of in vitro release from the intact tablets in water containing 60 % ethanol is shown in Figure 15A. and from crushed tablets in water containing 60 % ethanol is shown in Figure 15S.
Similar results were obtained when the water contained either 20 % or 40 % ethanol. The results show that alcohol concentrations up to at least 60 % have little or no effect on the release profiles. With respect to the crushed tablets, and as shown in Figure 15B, less than 25% of the Tramadol was released at 60 minutes in water containing 60% ethanol.
EXAMPLE 9¨ Exemplary Once-a-Day 200 mg Tramadol Tablet [00140] This Example describes the manufacture and testing of an exemplary once-a-day 200 mg tramadol HC1 tablet, where the tablets have a monolithic core and a controlled release coating. The core comprises super absorbent polycarbophil and the controlled release coat comprises xanth.an gum and Kollidon. Tramadol containing microparticles are disposed within the core and the coat.
[00141] The composition of four different lots of microparticles are set forth in Table 20.
Table 20 Ingredients. .% coliposition . = == . . =
= = ' LOT LOT 2 . LOT 3. LQT 4 . .
Tramado1HC1 58.3 57.4 69.6 69.6 MCC Avicel PH 101 25.0 24.6 17.4 17.4 Eudragit RS30D + 16.7 16.4 13.0 13.0 Plasacryl + Triethyl citrate Opadry IL white 1.6 Total 100.0 100.0 100.0 100.0 [00142] The formulations of uncoated microparticles were produced as follows.
Tramadol and Avicel PH 101 were mixed in a mixer for 3 minutes under low shear conditions.
The dry blend then was wetted under agitation in the same mixer by gradually adding water until a homogeneous wet mass suitable for extrusion was produced. The wet mass then was extruded at a constant speed (45 rpm) using a Laboratory Multigranulator extruder model MG-55 from LCI, Inc., NC, USA equipped with a dome die having a 0.6 min diameter hole and a fixed extrusion gap. The extrudates then were spheronized at a constant speed (1,800 rpm) using a Marumerzier Model QJ-230T from LCI, Inc., NC, USA. The wet microparticles were dried at 45 C in a fluid bed until a moisture content of about 2 % was achieved I001431 The resulting microparticles then were coated with an aqueous solution containing Eudragit RS 30D using a fluid bed coater. The microparticles were film coated to a weight gain of between 7% and 15%. Afterwards, for Lot 2 only, a curing solution containing Opadry 11 White was added to provide a film around the Eudragit containing coat.
[00144] The composition of the core granules is set forth in Table 21.
Table 21 Ingredients I % Composition Polycarbophil (Noveon AA-1) 80.00 MCC PI-I-101 19.50 Colloidal silicon dioxide 0.25 Sodium stearyl fumarate 0.25 Total 100.00 [00145] In addition to the controlled release microparticles, the core contained polycarbophilic acid as well as several other components. The remaining excipients for the core were mixed and subjected dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi. Then, the coated microparticles were mixed with the granulated core excipients to produce the core formulation [00146] The composition of the coat granules is set forth in Table 22, Ingreclienk % Composition Kollidon SR 312 Xanthan gum 66.3 Colloidal silicon dioxide 025 Sodium stearyl fumarate 0.25 Total 100 0 [0014'1 The remaining excipients for the coat were mixed and subjected to dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi. Then, coated microparticles were mixed with the granulated coat excipients to produce the coat formulation.
[00148] The composition of four different lots of tablets is set forth in Table 23.

Ingredients LOT 1 LOT 2 :O13:::.:::LOT 4.
%
Mg/tab J % Mg/tab % Mg/tab % MI/tab COM Compositions Coated Tramadol HC1 47.62 85.72 45.87 87.15 37.84 71.90 47.92 71.88 microparticles (50 mg Tramadol) Core granules 51.13 92.03 52.81 100.34 60.91 115.73 50 83 76 25 Colloidal silicon 0.50 0.90 0.50 0.95 0.50 0.95 0.50 - 0.75 dioxide Sodium story' 0.75 1.35 0.75 1.43 0.75 1.43 fumarate ___________________________________________________________ Total Core 100 180 100 190 100 190 100 150 Coat Compositions Coated Tramadol HCI 47.18 257.13 46.68 261.41 38.50 215.60 35.94 215.64 microparticles (150 mg Tramadol) Coat granules 51.82 282.42 52.32 292.99 52.13 292 04 63.06 378.36 Xanthan gum 8.35 46.76 --- ---Colloidal silicon 0.25 1.36 0.25 1.40 0.25 1.40 0.25 1.50 dioxide .
Sodium stearyl 0.75 4.09 0.75 4.20 0.75 4.20 0.75 4.50 fumarate ____________________________________________________________ Total Coat 100 545 100 560 100 560 100 600 Tablet Weight 725 750 750 750 [001491 Dry-coated tablets then were prepared using a Dry-Cota 16-Station tablet press from Manesty, UK. The core formulation was added to a first hopper in the tablet press and compressed into a core tablet. The coat formulation then was added to a second hopper in the tablet press and the core and the coat were compressed together to form the dry coated tablet.
The resulting dry coated tablets then were film coated with a solution of Opadry II using a frilly perforated pan coating machine (O'Hara, Mississauga, ON, CA).
[00150] The in vitro release properties of the resulting tablets (both intact and crushed) were measured in a U.S.P. Type I Apparatus in phosphate buffer 0-1 6.8 or water.
The results of in vitro release from intact tablets are shown in Figure 16A and from crushed tablets is shown in Figure 1613, The tablets were crushed using a pill crusher. The results show that the intact tablets of the invention demonstrated a controlled release of tramadol over 24 hours in phosphate buffer pH 6.8. Moreover, there was no dose dumping of tramadol from the crushed tablets when exposed to the same dissolution conditions. Under the conditions tested, less than 50 % of the tramadol was released within 60 minutes.
[00151] In addition, the in vitro release properties of the resulting tablets from Lot 4 (both intact and crushed) were measured in a U.S.P. Type I Apparatus in phosphate buffer pH
6.8 or water containing 20 % ethanol, 40 % ethanol and 60 % ethanol. The results of in vitro release from the intact tablets in buffer are shown in Figure 17A and from crushed tablets are shown in Figure 1713. The results show that alcohol concentrations up to at least 60 % have little or no effect on the release profiles_ With respect to the crushed tablets, less than 15% of the tramadol was released at 60 minutes in water containing 60% ethanol EXAMPLE 10¨ Exemplary Twelve Hour 30 mg Hydrocodone Bitartrate Tablet [00152] This Example describes the manufacture and testing of an exemplary twelve hour tablet containing 30 mg of Hydrocodone bitartrate. The tablets have a monolithic core and a controlled release coating. The core comprises super absorbent polycarbophil and the controlled release coat comprises xanthan gum and Kollidon. Hydrocodone containing microparticles are disposed within the coat. No active ingredient was disposed within the core [001531 The composition of the hydrocodone containing micropaiticles is set forth in Table 24.

Ingredients ____________________ . % Composition Hydrocodone bitarnate 31.82 MCC Avicel PH 101 59.09 , Eudragit RS301)10+ Plasacrxle Triethyl citrate 9.09 Total 100.00 [00154] The microparticles were produced as follows. Hydrocodone bitartrate and Avicel PH
101 were mixed in a mixer for 3 minutes under Low shear conditions. The dry blend then was wetted under agitation in the same mixer by gradually adding water until a homogeneous wet mass suitable for extrusion was produced. The wet mass then was extruded at a constant speed (45 rpm) using a Laboratory Multigranulator extruder model MG-55 from LCI, Inc., NC, USA
equipped with a dome die having a 0.6 mm diameter hole and a fixed extrusion gap. The extrudes then were spheronized at a constant speed (13800 rpm) using a Marumerzier Model QJ-230T from LCI, Inc., NC, USA. The wet microparticles were dried at 45 C in a fluid bed until a moisture content of about 2 % was achieved.
[001551 The resulting microparticles were coated with an aqueous solution containing Eudragit RS 30D using a fluid bed water The microparticles were film coated to a weight gain of between 7% and 15%.
(001561 The composition of the core granules is set forth in Table 25.

Ingredients . _____________ % CoMndsition Polycarbophil (Noveon AA-1) 80.00 =
MCC P14-101 19.50 Colloidal silicon dioxide 0.25 Sodium stearyl fumarate 0.25 Total 100.00 [00157] The core contained polycarbophil as well as several other components.
These excipients were mixed and subjected dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi.
[00158] The composition of the coat granules is set forth in Table 26.

IniredienTs. . . .% Composition .
Kollidon SR 33.17 Xanthan gum 66.33 Colloidal silicon dioxide 0.25 Sodium stearyl fumarate 0.25 Total 100.00 [001591 The remaining excipients for the coat were mixed and subjected to dry granulation in a roller compactor (Vector Corp.) under a roll speed of 5 rpm, a screw speed of 19 rpm, and a pressure of 800 psi. Then, the microparticles were mixed with the granulated coat excipients to produce the coat formulation.

[00160] The composition of intact tablets is set forth in Table 27.

....................................... Composition Ingredients % Mgt tab Cor- e Formulation - -Hydrocodone bitartrate microparticles Core granules 45 80 77.86 Klucel HF 52.95 90.02 Colloidal silicon dioxide 0.50 0.85 Sodium steely' fumarate 0.75 1.28 Total 100.00 170 00 Coat Formulation Hydrocodone bitaitrate micropartioies 21.93 94.30 Coat granules 53.81 231.38 Avicel PH 102 23.26 100.02 Colloidal silicon dioxide 0.25 1.08 Sodium stemyl fumaiate 0.75 ' 3.23 Total 100.00 430.00 [00161] Dry-coated tablets then were prepared using a Dry-Cota 16-Station tablet press from Manesty, UK. The core formulation was added to a first hopper in the tablet press and compressed into a core tablet. The coat formulation then was added to a second hopper in the tablet press and the core and the coat were compressed together to form the dry coated tablet.
The resulting dry coated tablets then were film coated with a solution of Opadry II using a fully perforated pan coating machine (O'Hara, Mississauga, ON, CA).
[00162] The in vitro release properties of the resulting tablets (both intact and crushed) were measured in a U.S.P. Type I Apparatus in phosphate buffer pH 6.8 or 0.1M
hydrochloric acid pH
1.2. The results of an vitro release from the intact tablets are shown in Figure 18A and from crushed tablets are shown in Figure 18B. The tablets were crushed by using a pill crusher. The results show that the intact tablets demonstrated a controlled release of Hydrocodone bitartrate over 12 hours in phosphate buffer pH 6.8 and in acid pH 1.2. However, the drug release rate in the acid was slightly higher than the release in phosphate buffer pH 6.8.
Furthermore, there was no dose dumping of hydrocodone bitartrate from the crushed tablets when exposed to the same dissolution conditions. Under the conditions tested, less than 30 %, and 55%
of hydrocodone was reic.ased within 60 minutes in phosphate buffer pH 6.8 and in acid pH 1.2, respectively, [00163] The scope of the claims should not be limited by the preferred embodiments set forth herein, but should be given the broadest interpretation consistent with the description as a whole.

Claims (91)

CLAIMS:
1. A solid, compressed controlled release formulation, comprising:
(a) a core comprising a superabsorbent material selected from the group consisting of an acrylic acid polymer cross-linked with divinyl glycol, an acrylic acid polymer cross-linked with allyl ethers of pentaerythritol and a mixture thereof, the superabsorbent material comprising from 30% to 70% (w/w) of the core;
(b) a controlled release coat surrounding the core; and (c) a plurality of controlled release microparticles having a pharmaceutically active agent disposed therein, wherein the microparticles are disposed within the core, the coat, or both the core and the coat, the formulation having a hardness from about 200 N
to about 400 N, and wherein the formulation (i) when intact and exposed to an aqueous medium, the pharmaceutically active agent is released from the formulation over a prolonged period of time, (ii) when crushed to break the controlled release coat and expose the core, and exposed to a volume of an aqueous medium that weighs 30 times the weight of the superabsorbent material in the formulation, the formulation absorbs all of the aqueous medium and swells to create a hard gel that traps the microparticles, and the microparticles provide controlled release of the pharmaceutically active agent, and (iii) when broken and exposed to 900 mL of water in a U.S.P. Type I
Apparatus with stirring at 100 rpm for 30 minutes at 37°C, less than about 50% by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the water.
2. The formulation of claim 1, wherein the microparticles are disposed within the core.
3. The formulation of claim 1, wherein the microparticles are disposed within the coat.
4. The formulation of claim 1, wherein the microparticles are disposed within both the core and the coat.
5. The formulation of any one of claims 1 to 4, wherein the core is monolithic.
6. The formulation of any one of claims 1 to 5, wherein the pharmaceutically active agent is released over a period of at least 12 hours.
7. The formulation of claim 6, wherein the pharmaceutically active agent is released over a period of at least 24 hours.
8. The formulation of any one of claims 1 to 7, wherein the superabsorbent material is present at about 30% to about 50% (w/w) of the core.
9. The formulation of any one of claims 1 to 8, wherein the core further comprises a granulating agent.
10. The formulation of claim 9, wherein the granulating agent is selected from the group consisting of xanthan gum, polyethylene oxide, polyvinylpyrollidone, cellulose and sucrose derivatives, and mixtures thereof.
11. The formulation of any one of claims 1 to 10, wherein the coat comprises a controlled release agent.
12. The formulation of claim 11, wherein the controlled release agent is selected from the group consisting of acetate succinate, a polyvinyl derivative, polyethylene oxide, polyacrylic acid, modified starch, cross-linked high amylose starch, hydroxypropyl starch, hydroxypropyl methylcellulose phthalate, cellulose, microcrystalline cellulose, carboxymethylethyl cellulose, cellulose acetate, methylcellulose, ethylcellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, cellulose phthalate, cellulose acetate, cellulose acetate phthalate, cellulose acetate propionate, cellulose acetate succinate, cellulose acetate butyrate, cellulose acetate trimellitate, poloxamer, povidone, alginic acid, sodium alginate, polyethylene glycol, polyethylene glycol alginate, gums, polymethacrylate, a copolymer of methacrylic acid and ethyl acrylate, a copolymer of polymethyl vinyl ether and malonic acid anhydride, a copolymer of polymethyl vinyl ether and malonic acid or the ethyl-, isopropyl-, n-butylesters thereof, zein, and mixtures of any of the foregoing.
13. The formulation of any one of claims 1 to 12, wherein the coat further comprises a viscosity-increasing agent.
14. The formulation of claim 13, wherein the viscosity-increasing agent is selected from the group consisting of xanthan gum, polyethylene oxide, polyvinylpyrollidone, cellulose and a sucrose derivative.
15. The formulation of any one of claims 1 to 14, wherein the microparticles comprise a controlled release agent.
16. The formulation of claim 15, wherein the controlled release agent is selected from the group consisting of acetate succinate, polyvinyl derivatives, polyethylene oxide, polyacrylic acid, modified starch, cross-linked high amylose starch, hydroxypropyl starch, hydroxypropyl methylcellulose phthalate, cellulose, microcrystalline cellulose, carboxymethylethyl cellulose, cellulose acetate, methylcellulose, ethylcellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, cellulose phthalate, cellulose acetate, cellulose acetate phthalate, cellulose acetate propionate, cellulose acetate succinate, cellulose acetate butyrate, cellulose acetate trimellitate, poloxamer, povidone, alginic acid, sodium alginate, polyethylene glycol, polyethylene glycol alginate, gums, polymethacrylate, a copolymer of methacrylic acid and ethyl acrylate, a copolymer of polymethyl vinyl ether and malonic acid anhydride, a copolymer of polymethyl vinyl ether and malonic acid or the ethyl-, isopropyl-, n-butylesters thereof, zein, and mixtures of any of the foregoing.
17. The formulation of any one of claims 1 to 16, wherein the microparticles are coated with a controlled release coat.
18. The formulation of claim 17, wherein the controlled release coat is selected from the group consisting of polymethacrylate, cellulose, a cellulose derivative, polyvinyl acetate, polyvinyl pyrollidone, and mixtures thereof.
19. The formulation of claim 18, wherein the cellulose derivative is selected from the group consisting of ethyl cellulose and hydroxypropyl cellulose.
20. The formulation of claim 18, wherein the polymethacrylate comprises a polymeric mixture of ethyl acrylate, methyl methacrylate and trimethylammonioethyl methacrylate.
21. The formulation of any one of claims 1 to 20, wherein the microparticles have an average diameter in the range of from about 1 µm to about 1000 µm.
22. The formulation of claim 21, wherein the microparticles have an average diameter in the range of from about 200 µm to about 900 µm.
23. The formulation of claim 22, wherein the microparticles have an average diameter in the range of from about 300 pm to about 800 um.
24. The formulation of claim 23, wherein the microparticles have an average diameter of about 700 µm.
25. The formulation of claim 21, wherein the microparticles have an average diameter in the range of from about 1 µm to about 400 µm.
26. The formulation of claim 25, wherein the microparticles have an average diameter in the range of from about 5 µm to about 300 µm.
27. The formulation of claim 26, wherein the microparticles have an average diameter in the range of from about 10 µm to about 200 µm.
28. The formulation of claim 27, wherein the microparticles have an average diameter of about 100 µm.
29. The formulation of any one of claims 1 to 28, wherein, in element (iii), less than about 25% by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the water.
30. The formulation of any one of claims 1 to 29, wherein, when the formulation is broken and exposed to 900 mL of an aqueous solution containing 60% (v/v) ethanol in a U.S.P. Type I Apparatus with stirring at 100 rpm for 30 minutes at 37°C, less than about 50% by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the aqueous solution.
31. The formulation of claim 30, wherein less than about 25% by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the aqueous solution.
32. The formulation of any one of claims 1 to 31, wherein the formulation is in the form of a capsule, caplet, pill, or a compressed tablet.
33. The formulation of any one of claims 1 to 32, wherein the pharmaceutically active agent is a drug capable of abuse.
34. The formulation of claim 33, wherein the drug is an opioid analgesic, hypnotic agent, anxiolytic, or a respiratory stimulant.
35. The formulation of claim 34, wherein the opioid analgesic is selected from the group consisting of alfentanil, buprenorphine, butorphanol, carefentanil, codeine, dezocine, diacetylmorphine, dihydrocodeine, dihydromorphine, diprenorphine, etorphine, fentanyl, hydrocodone, hydromorphone,13-hydroxy-3-methylfentanyl,levo a-acetylmethadol, levorphanol, lofentanil, meperidine, methadone, morphine, nalbuphine, oxycodone, oxymorphone, pentazocine, pethidine, prepoxyphene, remifentanil, sufentanil, tilidine, tramadol, and pharmaceutically acceptable salts, esters and prodrugs thereof.
36. The formulation of claim 34, wherein the hypnotic agent is selected from the group consisting of alprazolam, diazepam, flurazepam, loprazolam mexazolam, nitrazepam, barbiturate, chlormethiazole, eszopiclone, ramaelteon, zaleplon, zopiclone, zolpidem, and pharmaceutically acceptable salts, esters and prodrugs thereof.
37. The formulation of claim 34, wherein the anxiolytic is selected from the group consisting of amphetamine, alprazolan, diazepam, lorazepam, medazepam, oxazepam, pentylenetetrazole, and pharmaceutically acceptable salts, esters and prodrugs thereof.
38. The formulation of claim 34, wherein the respiratory stimulant is selected from the group consisting of caffeine, theophylline, amphetamine, benzphetamine hydrochloride, dextroamphetamine, dextroamphetamine sulfate, levamphetamine, levamphetamine hydrochloride, methamphetamine, methamphetamine hydrochloride, methylphenidate, methylphenidate hydrochloride, modafinil, pemoline, sibutramine, and sibutramine hydrochloride, and pharmaceutically acceptable salts, esters and prodrugs thereof.
39. The formulation of any one of claims 1 to 38, wherein the formulation, when crushed to break the controlled release coat and expose the core and exposed to 2 mL
of an aqueous medium, the formulation absorbs all of the aqueous medium and swells to create a hard gel that traps the microparticles.
40. The formulation of any one of claims 1 to 38, wherein the formulation, when crushed to break the controlled release coat and expose the core and exposed to 10 mL
of an aqueous medium, the formulation absorbs all of the aqueous medium and swells to create a hard gel that traps the microparticles.
41. The formulation of any one of claims 1 to 40, wherein the superabsorbent material is polycarbophil.
42. The formulation of any one of claims 1 to 40, wherein the superabsorbent material is polycarbophilic calcium.
43. The formulation of any one of claims 1 to 40, wherein the superabsorbent material is a carbomer homopolymer type A.
44. The formulation of any one of claims 1 to 40, wherein the superabsorbent material is a carbomer homopolymer type B.
45. A use of the controlled release formulation of any one of claims 1 to 44 for providing controlled release of the pharmaceutically active agent to an individual.
46. A use of the controlled release formulation of any one of claims 1 to 44 for preparation of a medicament for providing controlled release of the pharmaceutically active agent to an individual.
47. The controlled release formulation of any one of claims 1 to 44 for use in providing controlled release of the pharmaceutically active agent to an individual.
48. A solid, compressed controlled release formulation, comprising:
(a) a core comprising a superabsorbent material comprising a cross-linked acrylic acid polymer characterized in that 1 gram of the cross-linked acrylic acid polymer absorbs at least 30 mL of water, the superabsorbent material comprising from 30% to 70%
(w/w) of the core;
(b) a controlled release coat surrounding the core; and (c) a plurality of controlled release microparticles having a pharmaceutically active agent disposed therein, wherein the microparticles arc disposed within the core, the coat, or both the core and the coat, the formulation having a hardness from about 200 N to about 400 N, and wherein the formulation when intact and exposed to an aqueous medium, the pharmaceutically active agent is released from the formulation over a prolonged period of time, (i) when crushed to break the controlled release coat and expose the core, and exposed to a volume of an aqueous medium that weighs 30 times the weight of the superabsorbent material in the formulation, the formulation absorbs all of the aqueous medium and swells to create a hard gel that traps the microparticles, and the microparticles provide controlled release of the pharmaceutically active agent, and (ii) when broken and exposed to 900 mL of water in a U.S.P. Type I
Apparatus with stirring at 100 rpm for 30 minutes at 37°C, less than about 50% by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the water.
49. The formulation of claim 48, wherein the microparticles are disposed within the core.
50. The formulation of claim 48, wherein the microparticles are disposed within the coat.
51. The formulation of claim 48, wherein the microparticles are disposed within both the core and the coat.
52. The formulation of any one of claims 48 to 51, wherein the core is monolithic.
53. The formulation of any one of claims 48 to 52, wherein the core comprises a first layer, a second layer and an optional third layer.
54. The formulation of claim 53, wherein the first layer comprises the microparticles.
55. The formulation of any one of claims 48 to 54, wherein the pharmaceutically active agent is released over a period of at least 12 hours.
56. The formulation of claim 55, wherein the pharmaceutically active agent is released over a period of at least 24 hours.
57. The formulation of any one of claims 48 to 56, wherein the superabsorbent material is present at about 30% to about 50% (w/w) of the core.
58. The formulation of any one of claims 48 to 57, wherein the core further comprises a granulating agent.
59. The formulation of claim 58, wherein the granulating agent is selected from the group consisting of xanthan gum, polyethylene oxide, polyvinylpyroilidone, cellulose and sucrose derivatives, and mixtures thereof.
60. The formulation of any one of claims 48 to 59, wherein the coat comprises a controlled release agent.
61. The formulation of claim 60, wherein the controlled release agent is selected from the group consisting of acetate succinate, a polyvinyl derivative, polyethylene oxide, polyacrylic acid, modified starch, cross-linked high amylose starch, hydroxypropyl starch, hydroxypropyl methylcellulose phthalate, cellulose, microcrystalline cellulose, carboxymethylethyl cellulose, cellulose acetate, methylcellulose, ethylcellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, cellulose phthalate, cellulose acetate, cellulose acetate phthalate, cellulose acetate propionate, cellulose acetate succinate, cellulose acetate butyrate, cellulose acetate trimellitate, poloxamer, povidone, alginic acid, sodium alginate, polyethylene glycol, polyethylene glycol alginate, gums, polymethacrylate, a copolymer of methacrylic acid and ethyl acrylate, a copolymer of polymethyl vinyl ether and malonic acid anhydride, a copolymer of polymethyl vinyl ether and malonic acid or the ethyl-, isopropyl-, n-butylesters thereof, zein, and mixtures of any of the foregoing.
62. The formulation of any one of claims 48 to 61, wherein the coat further comprises a viscosity-increasing agent.
63. The formulation of claim 62, wherein the viscosity-increasing agent is selected from the group consisting of xanthan gum, polyethylene oxide, polyvinylpyrollidone, cellulose and a sucrose derivative.
64. The formulation of any one of claims 48 to 63, wherein the microparticles comprise a controlled release agent.
65. The formulation of claim 64, wherein the controlled release agent is selected from the group consisting of acetate suceinate, polyvinyl derivatives, polyethylene oxide, polyacrylic acid, modified starch, cross-linked high amylose starch, hydroxypropyl starch, hydroxypropyl methyleellulose phthalate, cellulose, microcrystalline cellulose, carboxymethylethyl cellulose, cellulose acetate, methylcellulose, ethylcellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, cellulose phthalate, cellulose acetate, cellulose acetate phthalate, cellulose acetate propionate, cellulose acetate succinate, cellulose acetate butyrate, cellulose acetate trimellitate, poloxamer, povidone, alginic acid, sodium alginate, polyethylene glycol, polyethylene glycol alginate, gums, polymethacrylate, a copolymer of methacrylic acid and ethyl acrylate, a copolymer of polymethyl vinyl ether and malonic acid anhydride, a copolymer of polymethyl vinyl ether and malonic acid or the ethyl-, isopropyl-, n-butylesters thereof, zein, and mixtures of any of the foregoing.
66. The formulation of any one of claims 48 to 65, wherein the microparticles are coated with a controlled release coat.
67. The formulation of claim 66, wherein the controlled release coat is selected from the group consisting of polymethacrylate, cellulose, a cellulose derivative, polyvinyl acetate, polyvinyl pyrollidone, and mixtures thereof.
68. The formulation of claim 67, wherein the cellulose derivative is selected from the group consisting of ethyl cellulose and hydroxypropyl cellulose.
69. The formulation of' claim 67, wherein the polymethacrylate comprises a polymeric mixture of ethyl acrylate, methyl methacrylate and trimethylammonioethyl methacrylate.
70. The formulation of any one of claims 48 to 69, wherein the microparticles have an average diameter in the range of from about 1 µm to about 1000 µm, from about 200 µm to about 900 µm, or from about 300 µm to about 800 µm.
71. The formulation of claim 70, wherein the microparticles have an average diameter of about 700 µm.
72. The formulation of claim 70, wherein the microparticles have an average diameter in the range of from about 1 µm to about 400 µm, from about 5 µm to about 300 µm, or from about 10 µm to about 200 µm.
73. The formulation of claim 72, wherein the microparticles have an average diameter of about 100 µm.
74. The formulation of any one of claims 48 to 73, wherein, in element (iii), less than about 25% by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the water.
75. The formulation of any one of claims 48 to 74, wherein, when the formulation is broken and exposed to 900 mL of an aqueous solution containing 60% (v/v) ethanol in a U.S.P. Type 1 Apparatus with stirring at 100 rpm for 30 minutes at 37°C, less than about 50%
by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the aqueous solution.
76. The formulation of claim 75, wherein less than about 25% by weight of the pharmaceutically active agent originally present in the formulation before it was broken is released into the aqueous solution.
77. The formulation of any one of claims 48 to 76, wherein the formulation is in the form of a capsule, caplet, pill, or a compressed tablet.
78. The formulation of any one of claims 48 to 77, wherein the pharmaceutically active agent is a drug capable of abuse.
79. The formulation of claim 78, wherein the drug is an opioid analgesic, hypnotic agent, anxiolytic, or a respiratory stimulant.
80. The formulation of any one of claims 48 to 79, wherein the formulation, when crushed to break the controlled release coat and expose the core and exposed to 2 mL
of an aqueous medium, the-formulation absorbs all of the aqueous medium and swells to create a hard gel that traps the microparticles.
81. The formulation of any one of claims 48 to 80, wherein the formulation, when crushed to break the controlled release coat and expose the core and exposed to 10 mL of an aqueous medium, the formulation absorbs all of the aqueous medium and swells to create a hard gel that traps the microparticles.
82. The formulation of any one of claims 48 to 81, wherein the cross-linked acrylic acid polymer is a copolymer.
83. The formulation of any one of claim 48 to 81, wherein the cross-linked acrylic acid polymer is selected from the group consisting of an acrylic acid polymer cross-linked with divinyl glycol, an acrylic acid polymer cross-linked with allyl ethers of pentaerythritol, and a mixture thereof.
84. The formulation of any one of claim 48 to 81, wherein the cross-linked acrylic acid polymer is polycarbophil.
85. The formulation of any one of claim 48 to 81, wherein the cross-linked acrylic acid polymer is polycarbophilic calcium.
86. The formulation of any one of claim 48 to 81, wherein the cross-linked acrylic acid polymer is a carbomer homopolymer type A.
87. The formulation of any one of claim 48 to 81, wherein the cross-linked acrylic acid polymer is a carbomer homopolymer type B.
88, The formulation of any one of claim 48 to 81, wherein 1 gram of the cross-linked acrylic acid polymer absorbs at least 50 mL of water.
89. A use of the controlled release formulation of any one of claims 48 to 88 for providing controlled release of the pharmaceutically active agent to an individual.
90. A use of the controlled release formulation of any one of claims 48 to 88 for preparation of a medicament for providing controlled release of the pharmaceutically active agent to an individual.
91. The controlled release formulation of any one of claims 48 to 88 for use in providing controlled release of the pharmaceutically active agent to an individual.
CA2707980A 2007-12-17 2008-12-16 Misuse preventative, controlled release formulation Active CA2707980C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US1429607P 2007-12-17 2007-12-17
US61/014,296 2007-12-17
PCT/CA2008/002200 WO2009076764A1 (en) 2007-12-17 2008-12-16 Misuse preventative, controlled release formulation

Publications (2)

Publication Number Publication Date
CA2707980A1 CA2707980A1 (en) 2009-06-25
CA2707980C true CA2707980C (en) 2015-05-12

Family

ID=40795147

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2707980A Active CA2707980C (en) 2007-12-17 2008-12-16 Misuse preventative, controlled release formulation

Country Status (13)

Country Link
US (7) US8486448B2 (en)
EP (1) EP2219612A4 (en)
JP (1) JP5651818B2 (en)
KR (1) KR20100121463A (en)
CN (1) CN101969930A (en)
AR (1) AR069905A1 (en)
AU (1) AU2008338207A1 (en)
BR (1) BRPI0821732A2 (en)
CA (1) CA2707980C (en)
MX (1) MX2010006694A (en)
RU (1) RU2010129907A (en)
TW (1) TW200934532A (en)
WO (1) WO2009076764A1 (en)

Families Citing this family (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030068375A1 (en) 2001-08-06 2003-04-10 Curtis Wright Pharmaceutical formulation containing gelling agent
US20040253310A1 (en) 2001-09-21 2004-12-16 Gina Fischer Morphine polymer release system
US20040234602A1 (en) 2001-09-21 2004-11-25 Gina Fischer Polymer release system
US7776314B2 (en) 2002-06-17 2010-08-17 Grunenthal Gmbh Abuse-proofed dosage system
ES2360102T3 (en) 2003-03-26 2011-05-31 Egalet A/S SYSTEM FOR CONTROLLED RELEASE OF MORPHINE.
US20070048228A1 (en) 2003-08-06 2007-03-01 Elisabeth Arkenau-Maric Abuse-proofed dosage form
DE10361596A1 (en) 2003-12-24 2005-09-29 Grünenthal GmbH Process for producing an anti-abuse dosage form
DE102005005446A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Break-resistant dosage forms with sustained release
DE10336400A1 (en) 2003-08-06 2005-03-24 Grünenthal GmbH Anti-abuse dosage form
US7201920B2 (en) 2003-11-26 2007-04-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of opioid containing dosage forms
DE102004032049A1 (en) 2004-07-01 2006-01-19 Grünenthal GmbH Anti-abuse, oral dosage form
DE102005005449A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Process for producing an anti-abuse dosage form
AU2008258596B2 (en) 2007-06-04 2013-02-14 Egalet Ltd Controlled release pharmaceutical compositions for prolonged effect
JP5651818B2 (en) 2007-12-17 2015-01-14 パラディン ラブス インコーポレーテッド Controlled release formulation to prevent misuse
EP2249811A1 (en) 2008-01-25 2010-11-17 Grünenthal GmbH Pharmaceutical dosage form
WO2009114648A1 (en) 2008-03-11 2009-09-17 Depomed Inc. Gastric retentive extended-release dosage forms comprising combinations of a non-opioid analgesic and an opioid analgesic
US8372432B2 (en) 2008-03-11 2013-02-12 Depomed, Inc. Gastric retentive extended-release dosage forms comprising combinations of a non-opioid analgesic and an opioid analgesic
EP2331629B1 (en) * 2008-09-15 2016-06-22 Paladin Labs Inc. Starch-based microparticles for the release of agents disposed therein
MY161550A (en) * 2008-10-27 2017-04-28 Alza Corp Extended release oral acetaminophen/tramadol dosage form
US8486449B2 (en) * 2008-12-16 2013-07-16 Paladin Labs Inc. Misuse preventative, controlled release formulation
NZ594207A (en) 2009-02-06 2013-03-28 Egalet Ltd Immediate release composition resistant to abuse by intake of alcohol
EP2445487A2 (en) 2009-06-24 2012-05-02 Egalet Ltd. Controlled release formulations
PE20120572A1 (en) 2009-07-22 2012-06-06 Gruenenthal Chemie HANDLING RESISTANT STABILIZED OXIDATION DOSAGE FORM
EP2456427B1 (en) 2009-07-22 2015-03-04 Grünenthal GmbH Hot-melt extruded controlled release dosage form
AU2010300641B2 (en) 2009-09-30 2016-03-17 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse
US9198861B2 (en) 2009-12-22 2015-12-01 Mallinckrodt Llc Methods of producing stabilized solid dosage pharmaceutical compositions containing morphinans
US8597681B2 (en) 2009-12-22 2013-12-03 Mallinckrodt Llc Methods of producing stabilized solid dosage pharmaceutical compositions containing morphinans
MX347753B (en) * 2010-02-24 2017-05-10 Cima Labs Inc Abuse-resistant formulations.
JP5819329B2 (en) 2010-03-09 2015-11-24 アルカーメス ファーマ アイルランド リミテッド Alcohol-resistant enteric pharmaceutical composition
US20120015031A1 (en) * 2010-07-14 2012-01-19 Grunenthal Gmbh Novel gastro-retentive dosage forms
PE20131126A1 (en) 2010-09-02 2013-10-21 Gruenenthal Chemie ALTERATION RESISTANT DOSAGE FORM INCLUDING AN ANIONIC POLYMER
NZ607392A (en) 2010-09-02 2015-03-27 Gruenenthal Chemie Tamper resistant dosage form comprising inorganic salt
US8741885B1 (en) 2011-05-17 2014-06-03 Mallinckrodt Llc Gastric retentive extended release pharmaceutical compositions
US8858963B1 (en) 2011-05-17 2014-10-14 Mallinckrodt Llc Tamper resistant composition comprising hydrocodone and acetaminophen for rapid onset and extended duration of analgesia
US9050335B1 (en) 2011-05-17 2015-06-09 Mallinckrodt Llc Pharmaceutical compositions for extended release of oxycodone and acetaminophen resulting in a quick onset and prolonged period of analgesia
US10702485B2 (en) 2011-07-09 2020-07-07 Syntrix Biosystems Inc. Compositions and methods for overcoming resistance to tramadol
DK2736497T3 (en) * 2011-07-29 2017-11-13 Gruenenthal Gmbh Shock-resistant tablet that provides an immediate release of a drug.
EP2736495B1 (en) 2011-07-29 2017-08-23 Grünenthal GmbH Tamper-resistant tablet providing immediate drug release
FR2983409B1 (en) * 2011-12-06 2013-12-27 Ethypharm Sa COMPRESSOR CAPABLE OF COMBATTING INJECTION MISTAKE
MX356421B (en) 2012-02-28 2018-05-29 Gruenenthal Gmbh Tamper-resistant dosage form comprising pharmacologically active compound and anionic polymer.
MX356111B (en) 2012-04-18 2018-05-15 SpecGx LLC Immediate release, abuse deterrent pharmaceutical compositions.
LT2838512T (en) 2012-04-18 2018-11-12 GrĆ¼nenthal GmbH Tamper resistant and dose-dumping resistant pharmaceutical dosage form
US10064945B2 (en) 2012-05-11 2018-09-04 Gruenenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
EP2877161A1 (en) 2012-07-06 2015-06-03 Egalet Ltd. Abuse deterrent pharmaceutical compositions for controlled release
EP2872121B1 (en) 2012-07-12 2018-09-05 SpecGx LLC Extended release, abuse deterrent pharmaceutical compositions
KR102127625B1 (en) * 2012-09-03 2020-06-29 다이이찌 산쿄 가부시키가이샤 Hydromorphone hydrochloride-containing oral sustained-release pharmaceutical composition
CA2791206A1 (en) * 2012-09-28 2014-03-28 Pharmascience Inc. Abuse deterrent pharmaceutical formulation
US9101636B2 (en) 2012-11-30 2015-08-11 Acura Pharmaceuticals, Inc. Methods and compositions for self-regulated release of active pharmaceutical ingredient
BR112015021002B8 (en) 2013-03-15 2023-03-28 Mallinckrodt Llc SOLID PHARMACEUTICAL DOSAGE FORM COMPRISING AN ACTIVE PHARMACEUTICAL INGREDIENT
MX2015016254A (en) 2013-05-29 2016-04-20 Gruenenthal Gmbh Tamper resistant dosage form with bimodal release profile.
US10154966B2 (en) 2013-05-29 2018-12-18 Grünenthal GmbH Tamper-resistant dosage form containing one or more particles
DE102013009114A1 (en) 2013-05-29 2014-12-04 Franz Gerstheimer Pharmaceutical composition to overcome metabolic problems
KR20160031526A (en) 2013-07-12 2016-03-22 그뤼넨탈 게엠베하 Tamper-resistant dosage form containing ethylene-vinyl acetate polymer
CA3042642A1 (en) 2013-08-12 2015-02-19 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
CN105611915B (en) * 2013-08-14 2021-05-11 赢创运营有限公司 Coating composition
US9770514B2 (en) 2013-09-03 2017-09-26 ExxPharma Therapeutics LLC Tamper-resistant pharmaceutical dosage forms
CA2931553C (en) 2013-11-26 2022-01-18 Grunenthal Gmbh Preparation of a powdery pharmaceutical composition by means of cryo-milling
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
AU2015237723B2 (en) 2014-03-26 2018-04-26 Sun Pharma Advanced Research Company Ltd. Abuse deterrent immediate release biphasic matrix solid dosage form
EP3142646A1 (en) 2014-05-12 2017-03-22 Grünenthal GmbH Tamper resistant immediate release capsule formulation comprising tapentadol
US9872835B2 (en) 2014-05-26 2018-01-23 Grünenthal GmbH Multiparticles safeguarded against ethanolic dose-dumping
US11617712B2 (en) 2014-07-03 2023-04-04 SpecGx LLC Abuse deterrent immediate release formulations comprising non-cellulose polysaccharides
CA2910865C (en) 2014-07-15 2016-11-29 Isa Odidi Compositions and methods for reducing overdose
EP3169315B1 (en) 2014-07-17 2020-06-24 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US9132096B1 (en) 2014-09-12 2015-09-15 Alkermes Pharma Ireland Limited Abuse resistant pharmaceutical compositions
AU2015336065A1 (en) 2014-10-20 2017-05-04 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
WO2016170097A1 (en) 2015-04-24 2016-10-27 Grünenthal GmbH Tamper-resistant dosage form with immediate release and resistance against solvent extraction
WO2017042325A1 (en) 2015-09-10 2017-03-16 Grünenthal GmbH Protecting oral overdose with abuse deterrent immediate release formulations
US9849125B1 (en) 2015-11-03 2017-12-26 Banner Lifie Sciences LLC Anti-overingestion dosage forms
US10335375B2 (en) 2017-05-30 2019-07-02 Patheon Softgels, Inc. Anti-overingestion abuse deterrent compositions
GB2567493B (en) * 2017-10-13 2019-12-18 Altus Formulation Inc Starch-based release modifying excipients and pharmaceutical compositions derived therefrom
WO2019089330A1 (en) * 2017-11-01 2019-05-09 Edgemont Pharmceuticals, Llc Trust Alcohol-resistant oral pharmaceutical compositions of lorazepam
MX2021002459A (en) 2018-09-25 2021-04-29 SpecGx LLC Abuse deterrent immediate release capsule dosage forms.
WO2020086673A1 (en) * 2018-10-26 2020-04-30 Guangzhou Dazhou Biomedicine Ltd. Ketamine oral transmucosal delivery system
US11000488B2 (en) 2019-03-22 2021-05-11 Syntrix Biosystems Inc. Treating pain using desmetramadol
US20230241582A1 (en) * 2019-12-20 2023-08-03 Lg Chem, Ltd. Super Absorbent Polymer Composition
EP3943538B1 (en) * 2019-12-20 2023-08-23 Lg Chem, Ltd. Preparation method of super absorbent polymer composition
BR112021023200A2 (en) * 2019-12-20 2022-07-26 Lg Chemical Ltd METHOD OF PREPARATION OF SUPERABSORBENT POLYMER COMPOSITION
US20220242990A1 (en) * 2020-01-20 2022-08-04 Lg Chem, Ltd. Preparation method of super absorbent polymer
CN115702888A (en) * 2021-08-13 2023-02-17 合肥立方制药股份有限公司 Oxycodone hydrochloride osmotic pump sustained-release tablet and preparation method thereof
CN115707455A (en) * 2021-08-18 2023-02-21 越洋医药开发(广州)有限公司 Tablet allowing sleep regulation type drug to be released in stages and preparation method thereof
US20230120697A1 (en) * 2021-09-16 2023-04-20 Soane Materials Llc Swellable polymeric materials and useful articles incorporating same

Family Cites Families (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3004613A (en) 1956-05-17 1961-10-17 Milburn R Simmons Electronic deep hole condition analyser
US3773995A (en) 1972-10-27 1973-11-20 Westinghouse Electric Corp Motor advanced spring charging pawl and ratchet mechanism with spring assist
US3966940A (en) 1973-11-09 1976-06-29 Bristol-Myers Company Analgetic compositions
DE2530563C2 (en) * 1975-07-09 1986-07-24 Bayer Ag, 5090 Leverkusen Analgesic drugs with reduced potential for abuse
US5004613A (en) 1987-07-27 1991-04-02 Mcneil-Ppc, Inc. Oral sustained release pharmaceutical formulation and process
US4820522A (en) 1987-07-27 1989-04-11 Mcneilab, Inc. Oral sustained release acetaminophen formulation and process
US4968509A (en) 1987-07-27 1990-11-06 Mcneilab, Inc. Oral sustained release acetaminophen formulation and process
US5133974A (en) 1989-05-05 1992-07-28 Kv Pharmaceutical Company Extended release pharmaceutical formulations
JP2772695B2 (en) * 1990-01-26 1998-07-02 日水製薬株式会社 3-layer tablet
CA2041774C (en) 1990-11-27 1994-04-19 Mircea A. Mateescu Use of cross-linked amylose as a matrix for the slow release of biologically active compounds
US5603956A (en) 1990-11-27 1997-02-18 Labopharm Inc. Cross-linked enzymatically controlled drug release
KR100221695B1 (en) * 1991-08-12 1999-09-15 그린 마틴, 브라이언 쥐 테슬리 Pharmaceutical spheroid formulation
CA2095523C (en) 1991-09-06 2004-06-22 Robert B. Raffa Composition comprising a tramadol material and acetaminophen and its use
US5478577A (en) 1993-11-23 1995-12-26 Euroceltique, S.A. Method of treating pain by administering 24 hour oral opioid formulations exhibiting rapid rate of initial rise of plasma drug level
US5616343A (en) 1993-03-25 1997-04-01 Labopharm, Inc. Cross-linked amylose as a binder/disintegrant in tablets
IL119660A (en) 1993-05-10 2002-09-12 Euro Celtique Sa Controlled release formulation comprising tramadol
IL110014A (en) 1993-07-01 1999-11-30 Euro Celtique Sa Solid controlled-release oral dosage forms of opioid analgesics
IT1264696B1 (en) 1993-07-09 1996-10-04 Applied Pharma Res PHARMACEUTICAL FORMS INTENDED FOR ORAL ADMINISTRATION ABLE TO RELEASE ACTIVE SUBSTANCES AT A CONTROLLED AND DIFFERENTIATED SPEED
DE4329794C2 (en) 1993-09-03 1997-09-18 Gruenenthal Gmbh Tramadol salt-containing drugs with delayed release
US6210714B1 (en) 1993-11-23 2001-04-03 Euro-Celtique S.A. Immediate release tablet cores of acetaminophen having sustained-release coating
GB9401894D0 (en) 1994-02-01 1994-03-30 Rhone Poulenc Rorer Ltd New compositions of matter
US6548084B2 (en) * 1995-07-20 2003-04-15 Smithkline Beecham Plc Controlled release compositions
GB9519363D0 (en) 1995-09-22 1995-11-22 Euro Celtique Sa Pharmaceutical formulation
AUPN605795A0 (en) 1995-10-19 1995-11-09 F.H. Faulding & Co. Limited Analgesic pharmaceutical composition
US5773031A (en) 1996-02-27 1998-06-30 L. Perrigo Company Acetaminophen sustained-release formulation
US6159501A (en) 1996-03-08 2000-12-12 Nycomed Danmark A/S Modified release multiple-units dosage composition for release of opioid compounds
WO1997033566A2 (en) 1996-03-12 1997-09-18 Alza Corporation Composition and dosage form comprising opioid antagonist
US20040024006A1 (en) 1996-05-06 2004-02-05 Simon David Lew Opioid pharmaceutical compositions
BE1011045A3 (en) 1997-03-14 1999-04-06 Ucb Sa Pharmaceutical composition for controlled release of active substances.
ES2248908T7 (en) 1997-06-06 2014-11-24 Depomed, Inc. Dosage forms of drugs orally and gastric retention for continued release of highly soluble drugs
US6635280B2 (en) 1997-06-06 2003-10-21 Depomed, Inc. Extending the duration of drug release within the stomach during the fed mode
US20020054911A1 (en) * 2000-05-11 2002-05-09 Boehringer Mannheim Pharmaceutical Corporation-Sm Ithkline Beckman Corporation, Limited Partnershi Novel oral dosage form for carvedilol
CN1204890C (en) 1997-12-22 2005-06-08 欧罗赛铁克股份有限公司 Method for preventing abuse of opioid dosage forms
RU2241458C2 (en) 1997-12-22 2004-12-10 Эро-Селтик, С.А. Combinations of agonist/antagonist for opioid
FR2772615B1 (en) 1997-12-23 2002-06-14 Lipha MULTILAYER TABLET FOR INSTANT RELEASE THEN PROLONGED ACTIVE SUBSTANCES
US6284273B1 (en) 1998-02-24 2001-09-04 Vincent Lenaerts Cross-linked high amylose starch resistant to amylase as a matrix for the slow release of biologically active compounds
US6245357B1 (en) 1998-03-06 2001-06-12 Alza Corporation Extended release dosage form
US6156342A (en) 1998-05-26 2000-12-05 Andex Pharmaceuticals, Inc. Controlled release oral dosage form
US6245387B1 (en) 1998-11-03 2001-06-12 Diamon-Fusion International, Inc. Capped silicone film and method of manufacture thereof
ES2255490T3 (en) 1999-03-31 2006-07-01 Janssen Pharmaceutica N.V. PREGELATINIZED ALMIDON IN A CONTROLLED RELEASE FORMULATION.
US6306425B1 (en) 1999-04-09 2001-10-23 Southern Research Institute Injectable naltrexone microsphere compositions and their use in reducing consumption of heroin and alcohol
US20030118641A1 (en) 2000-07-27 2003-06-26 Roxane Laboratories, Inc. Abuse-resistant sustained-release opioid formulation
AU2276801A (en) 1999-12-20 2001-07-03 Schering Corporation Extended release oral dosage composition
JP2003522144A (en) 2000-02-08 2003-07-22 ユーロ−セルティーク,エス.エイ. Controlled release compositions comprising opioid agonists and antagonists
US6761895B2 (en) 2000-04-17 2004-07-13 Yamanouchi Pharmaceutical Co., Ltd. Drug delivery system for averting pharmacokinetic drug interaction and method thereof
GB0009522D0 (en) 2000-04-19 2000-06-07 Smithkline Beecham Plc Composition
US6955821B2 (en) 2000-04-28 2005-10-18 Adams Laboratories, Inc. Sustained release formulations of guaifenesin and additional drug ingredients
WO2001085282A1 (en) 2000-05-10 2001-11-15 Armstrong Brad A Analog controls for coffee makers
US6488962B1 (en) 2000-06-20 2002-12-03 Depomed, Inc. Tablet shapes to enhance gastric retention of swellable controlled-release oral dosage forms
US6607748B1 (en) 2000-06-29 2003-08-19 Vincent Lenaerts Cross-linked high amylose starch for use in controlled-release pharmaceutical formulations and processes for its manufacture
US20070060500A1 (en) 2000-08-22 2007-03-15 New River Pharmaceuticals Inc. Pharmaceutical compositions for prevention of overdose or abuse
CN101317825A (en) 2000-10-30 2008-12-10 欧罗赛铁克股份有限公司 Controlled release hydrocodone formulations
US20020187192A1 (en) 2001-04-30 2002-12-12 Yatindra Joshi Pharmaceutical composition which reduces or eliminates drug abuse potential
CN1525851A (en) 2001-05-11 2004-09-01 ������ҩ�����޹�˾ Abuse-resistant controlled-release opioid dosage form
US20030004177A1 (en) 2001-05-11 2003-01-02 Endo Pharmaceuticals, Inc. Abuse-resistant opioid dosage form
US20030064122A1 (en) 2001-05-23 2003-04-03 Endo Pharmaceuticals, Inc. Abuse resistant pharmaceutical composition containing capsaicin
US6968551B2 (en) 2001-06-11 2005-11-22 John Hediger System and user interface for generation and processing of software application installation instructions
US7141250B2 (en) 2001-08-06 2006-11-28 Euro-Celtique S.A. Pharmaceutical formulation containing bittering agent
US20030044458A1 (en) 2001-08-06 2003-03-06 Curtis Wright Oral dosage form comprising a therapeutic agent and an adverse-effect agent
ES2326794T3 (en) 2001-08-06 2009-10-20 Euro-Celtique S.A. FORMULATIONS OF OPIOID AGONISTS WITH LIBERABLE AND SEQUESTED ANTAGONISTS.
BR0212020A (en) 2001-08-06 2005-08-16 Euro Celtique Sa Dosage forms, methods for preventing abuse of a dosage form, methods for preventing diversion of a dosage form, methods for treating pain and method of preparing a dosage form
US7332182B2 (en) 2001-08-06 2008-02-19 Purdue Pharma L.P. Pharmaceutical formulation containing opioid agonist, opioid antagonist and irritant
US20030068375A1 (en) 2001-08-06 2003-04-10 Curtis Wright Pharmaceutical formulation containing gelling agent
US7157103B2 (en) 2001-08-06 2007-01-02 Euro-Celtique S.A. Pharmaceutical formulation containing irritant
KR20040060917A (en) 2001-08-06 2004-07-06 유로-셀티크 소시에떼 아노뉨 Compositions and methods to prevent abuse of opioids
US7144587B2 (en) 2001-08-06 2006-12-05 Euro-Celtique S.A. Pharmaceutical formulation containing opioid agonist, opioid antagonist and bittering agent
US7842307B2 (en) 2001-08-06 2010-11-30 Purdue Pharma L.P. Pharmaceutical formulation containing opioid agonist, opioid antagonist and gelling agent
US7375082B2 (en) 2002-02-22 2008-05-20 Shire Llc Abuse-resistant hydrocodone compounds
US7338939B2 (en) 2003-09-30 2008-03-04 New River Pharmaceuticals Inc. Abuse-resistant hydrocodone compounds
US7169752B2 (en) 2003-09-30 2007-01-30 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US20030049317A1 (en) 2001-08-30 2003-03-13 Lindsay David R. Method and composition for reducing the danger and preventing the abuse of controlled release pharmaceutical formulations
US20030092724A1 (en) 2001-09-18 2003-05-15 Huaihung Kao Combination sustained release-immediate release oral dosage forms with an opioid analgesic and a non-opioid analgesic
CA2459976A1 (en) 2001-09-26 2003-04-03 Penwest Pharmaceuticals Company Opioid formulations having reduced potential for abuse
US6723340B2 (en) 2001-10-25 2004-04-20 Depomed, Inc. Optimal polymer mixtures for gastric retentive tablets
US20040126428A1 (en) 2001-11-02 2004-07-01 Lyn Hughes Pharmaceutical formulation including a resinate and an aversive agent
CA2464528A1 (en) 2001-11-02 2003-05-15 Elan Corporation, Plc Pharmaceutical composition
US20030191147A1 (en) 2002-04-09 2003-10-09 Barry Sherman Opioid antagonist compositions and dosage forms
AU2003228654A1 (en) 2002-04-29 2003-11-17 The General Hospital Corporation Compositions and methods for preventing abuse of orally administered medications
CA2486075A1 (en) 2002-05-13 2003-11-20 Endo Pharmaceuticals Inc. Abuse-resistant opioid solid dosage form
WO2004041154A2 (en) 2002-05-13 2004-05-21 Endo Pharmaceuticals Inc. Abuse-resistant opioid dosage form
US7041320B1 (en) 2002-05-31 2006-05-09 Biotek, Inc. High drug loaded injectable microparticle compositions and methods of treating opioid drug dependence
US7776314B2 (en) 2002-06-17 2010-08-17 Grunenthal Gmbh Abuse-proofed dosage system
KR100522239B1 (en) 2002-07-16 2005-10-18 주식회사 서울제약 Compositions for controlled release acetaminophen dosage forms
PT1551372T (en) 2002-09-20 2018-07-23 Alpharma Pharmaceuticals Llc Sequestering subunit and related compositions and metohds
WO2004026308A1 (en) 2002-09-21 2004-04-01 Shuyi Zhang Sustained release compound of acetamidophenol and tramadol
JP5189242B2 (en) 2002-09-23 2013-04-24 アルケルメス ファーマ アイルランド リミテッド Abuse-resistant pharmaceutical composition
US20050191244A1 (en) 2002-10-25 2005-09-01 Gruenenthal Gmbh Abuse-resistant pharmaceutical dosage form
TWI319713B (en) 2002-10-25 2010-01-21 Sustained-release tramadol formulations with 24-hour efficacy
US8487002B2 (en) 2002-10-25 2013-07-16 Paladin Labs Inc. Controlled-release compositions
CA2507522C (en) 2002-12-13 2015-02-24 Durect Corporation Oral drug delivery system
CA2414500A1 (en) 2002-12-17 2004-06-17 Purepharm Inc. Agonist-aversive combination medicines
ES2371462T3 (en) 2003-01-13 2012-01-03 Shire Llc CONJUGATES OF CARBOHYDRATES TO PREVENT THE ABUSE OF CONTROLLED SUBSTANCES.
US20040202717A1 (en) 2003-04-08 2004-10-14 Mehta Atul M. Abuse-resistant oral dosage forms and method of use thereof
MY135852A (en) 2003-04-21 2008-07-31 Euro Celtique Sa Pharmaceutical products
US8906413B2 (en) 2003-05-12 2014-12-09 Supernus Pharmaceuticals, Inc. Drug formulations having reduced abuse potential
GB2403711A (en) 2003-07-07 2005-01-12 Gw Pharma Ltd Drug dispenser with controlled access
DE10336400A1 (en) 2003-08-06 2005-03-24 Grünenthal GmbH Anti-abuse dosage form
CN100588391C (en) 2003-08-12 2010-02-10 恩德制药公司 Single long acting slow-release tablet for deterring abuse of medicine
US20050048115A1 (en) 2003-08-27 2005-03-03 Murty Mangena Buprenorphine microspheres
US20060194826A1 (en) 2003-09-25 2006-08-31 Euro-Celtique S.A. Pharmaceutical combinations of hydrocodone and naltrexone
MXPA06003450A (en) 2003-09-26 2006-08-31 Johnson & Johnson Drug coating providing high drug loading and methods for providing the same.
US20050158382A1 (en) 2003-09-26 2005-07-21 Evangeline Cruz Controlled release formulations of opioid and nonopioid analgesics
US20080031901A1 (en) 2004-09-24 2008-02-07 Abbott Laboratories Sustained release monoeximic formulations of opioid and nonopioid analgesics
US20060172006A1 (en) 2003-10-10 2006-08-03 Vincent Lenaerts Sustained-release tramadol formulations with 24-hour clinical efficacy
US20060009478A1 (en) 2003-10-15 2006-01-12 Nadav Friedmann Methods for the treatment of back pain
EP2264864B1 (en) 2003-11-13 2019-10-02 SEW-EURODRIVE GmbH & Co. KG Compact drive
US7201920B2 (en) 2003-11-26 2007-04-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of opioid containing dosage forms
EP1691892B1 (en) 2003-12-09 2007-02-28 Euro-Celtique S.A. Tamper resistant co-extruded dosage form containing an active agent and an adverse agent and process of making same
BRPI0508769A (en) 2004-03-30 2007-08-28 Euro Celtique Sa tamper-resistant dosage form comprising an adsorbent and an adverse agent
EP1584335A3 (en) 2004-04-05 2006-02-22 Laboratorios Del Dr. Esteve, S.A. Active substance combination comprising a carbinol composition and an opioid
US20070224269A1 (en) * 2004-06-10 2007-09-27 Rubino Orapin P Controlled Release Pharmaceutical Formulation
DE102004032049A1 (en) * 2004-07-01 2006-01-19 Grünenthal GmbH Anti-abuse, oral dosage form
US8541026B2 (en) 2004-09-24 2013-09-24 Abbvie Inc. Sustained release formulations of opioid and nonopioid analgesics
US20060110327A1 (en) 2004-11-24 2006-05-25 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
US20070231268A1 (en) 2004-11-24 2007-10-04 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
FR2878158B1 (en) 2004-11-24 2009-01-16 Flamel Technologies Sa ORAL PHARMACEUTICAL FORM, SOLID MICROPARTICULAR DESIGNED TO PREVENT MEASUREMENT
US20060177380A1 (en) 2004-11-24 2006-08-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
US20080152595A1 (en) 2004-11-24 2008-06-26 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
FR2881652B1 (en) 2005-02-08 2007-05-25 Flamel Technologies Sa MICROPARTICULAR ORAL PHARMACEUTICAL FORM ANTI-MEASURING
FR2889810A1 (en) 2005-05-24 2007-02-23 Flamel Technologies Sa ORAL MEDICINAL FORM, MICROPARTICULAR, ANTI-MEASUREMENT
EP1695700A1 (en) 2005-02-28 2006-08-30 Euro-Celtique S.A. Dosage form containing oxycodone and naloxone
US7191215B2 (en) 2005-03-09 2007-03-13 Marquee, Inc. Method and system for providing instantaneous media-on-demand services by transmitting contents in pieces from client machines
WO2006110642A2 (en) 2005-04-07 2006-10-19 Hythiam, Inc. Improved methods of and compositions for the prevention of anxiety, substance abuse, and dependence
US20060257473A1 (en) 2005-05-11 2006-11-16 Porranee Puranajoti Extended release tablet
WO2006133733A1 (en) 2005-06-13 2006-12-21 Flamel Technologies Oral dosage form comprising an antimisuse system
US20070020339A1 (en) 2005-07-20 2007-01-25 Pharmorx Inc. Compositions and methods for controlling abuse of medications
PT1931315E (en) 2005-08-24 2014-01-03 Endo Pharmaceuticals Inc Sustained release formulations of nalbuphine
CA2616416A1 (en) 2005-09-09 2007-05-03 Labopharm, Inc. Trazodone composition for once a day adminisitiation
US8652529B2 (en) 2005-11-10 2014-02-18 Flamel Technologies Anti-misuse microparticulate oral pharmaceutical form
JP2009523833A (en) 2006-01-21 2009-06-25 アボット ゲーエムベーハー ウント カンパニー カーゲー Formulations and methods for drug delivery
US20070212414A1 (en) 2006-03-08 2007-09-13 Penwest Pharmaceuticals Co. Ethanol-resistant sustained release formulations
EP1993519A4 (en) 2006-03-15 2011-12-21 Acura Pharmaceuticals Inc Methods and compositions for deterring abuse of orally administered pharmaceutical products
JP4953673B2 (en) * 2006-03-22 2012-06-13 リンテック株式会社 Oral administration
US20070237816A1 (en) 2006-04-06 2007-10-11 David Finkelstein Acetaminophen formulation for joint pain relief
US9023400B2 (en) 2006-05-24 2015-05-05 Flamel Technologies Prolonged-release multimicroparticulate oral pharmaceutical form
WO2008011596A2 (en) 2006-07-21 2008-01-24 Lab International Srl Hydrophilic abuse deterrent delivery system
US8445018B2 (en) 2006-09-15 2013-05-21 Cima Labs Inc. Abuse resistant drug formulation
JP5651818B2 (en) * 2007-12-17 2015-01-14 パラディン ラブス インコーポレーテッド Controlled release formulation to prevent misuse
US8486449B2 (en) 2008-12-16 2013-07-16 Paladin Labs Inc. Misuse preventative, controlled release formulation

Also Published As

Publication number Publication date
AR069905A1 (en) 2010-03-03
KR20100121463A (en) 2010-11-17
AU2008338207A1 (en) 2009-06-25
EP2219612A1 (en) 2010-08-25
US20140294954A1 (en) 2014-10-02
US20140193494A1 (en) 2014-07-10
CN101969930A (en) 2011-02-09
US20160367483A1 (en) 2016-12-22
US20090175937A1 (en) 2009-07-09
WO2009076764A1 (en) 2009-06-25
US8486448B2 (en) 2013-07-16
MX2010006694A (en) 2010-09-07
US20150272890A1 (en) 2015-10-01
CA2707980A1 (en) 2009-06-25
US8691270B2 (en) 2014-04-08
TW200934532A (en) 2009-08-16
US8920834B2 (en) 2014-12-30
EP2219612A4 (en) 2013-10-30
RU2010129907A (en) 2012-01-27
JP5651818B2 (en) 2015-01-14
JP2011506493A (en) 2011-03-03
US8920833B2 (en) 2014-12-30
BRPI0821732A2 (en) 2015-06-16
US20130344142A1 (en) 2013-12-26
US20180185292A1 (en) 2018-07-05

Similar Documents

Publication Publication Date Title
CA2707980C (en) Misuse preventative, controlled release formulation
US8927014B2 (en) Misuse preventative, controlled release formulation
JP5635082B2 (en) Dosage form
JP5323134B2 (en) Sustained release gel coating composition
JP5965583B2 (en) Abuse resistant pharmaceutical composition, method of use and preparation
CA2686964A1 (en) Pharmaceutical formulation for the production of rapidly disintegrating tablets
EA036444B1 (en) Modified release abuse deterrent dosage forms
CA2970065A1 (en) Immediate release abuse-deterrent granulated dosage forms
CN111465390A (en) Modified release abuse deterrent dosage forms

Legal Events

Date Code Title Description
EEER Examination request