WO2008033351A2 - Multimodal abuse resistant and extended release formulations - Google Patents

Multimodal abuse resistant and extended release formulations Download PDF

Info

Publication number
WO2008033351A2
WO2008033351A2 PCT/US2007/019745 US2007019745W WO2008033351A2 WO 2008033351 A2 WO2008033351 A2 WO 2008033351A2 US 2007019745 W US2007019745 W US 2007019745W WO 2008033351 A2 WO2008033351 A2 WO 2008033351A2
Authority
WO
WIPO (PCT)
Prior art keywords
dosage form
hours
administration
less
drug
Prior art date
Application number
PCT/US2007/019745
Other languages
French (fr)
Other versions
WO2008033351A3 (en
Inventor
Najib Babul
Original Assignee
Theraquest Biosciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Theraquest Biosciences, Inc. filed Critical Theraquest Biosciences, Inc.
Publication of WO2008033351A2 publication Critical patent/WO2008033351A2/en
Publication of WO2008033351A3 publication Critical patent/WO2008033351A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/485Inorganic compounds
    • 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
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • 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
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds

Definitions

  • the present invention is in the field of multimodal oral, abuse resistant pharmaceutical compositions of abusable drugs, extended release pharmaceutical compositions of abusable drugs, extended release abuse resistant pharmaceutical compositions of abusable drugs and the use thereof.
  • a wide variety of pharmaceutical products can be subjected to abuse including psychostimulants, amphetamine and amphetamine analogs, amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, barbiturates and cannabinoid agonists.
  • Some of the abusable substances within these classes of drugs are currently available for commercial sale to prevent or treat medical conditions, while others are in development.
  • opioids are abused or used for nonmedical reasons can alter brain activity and lead to dependence.
  • Commonly abused classes of prescription drugs include opioids (often prescribed to treat pain), central nervous system depressants (often prescribed to treat anxiety and sleep disorders), and stimulants (prescribed to treat narcolepsy, ADHD, and obesity).
  • Commonly used opioids include oxycodone (OxyContinTM), propoxyphene (DarvonTM), hydrocodone (Vicodin), hydromorphone (DilaudidTM), meperidine (DemerolTM), and diphenoxylate (LomotilTM).
  • Common central nervous system depressants include barbiturates such as pentobarbital sodium (NembutalTM), and benzodiazepines such as diazepam (ValiumTM) and alprazolam (XanaxTM). Stimulants include dextroamphetamine (DexedrineTM) and methylphenidate (RitalinTM). Long- term use of opioids or central nervous system depressants can lead to physical dependence and addiction. Taken in high doses, stimulants can lead to compulsive use, paranoia, dangerously high body temperatures, and irregular heartbeat.” (www.nida.nih.gov, accessed September 7, 2006)
  • THC delta-9- tetrahydrocannabinol
  • Marijuana is the most commonly used illicit drug in the United States.
  • the Drug Abuse Warning Network (DAWN), a system for monitoring the health impact of drugs, estimated that, in 2002, marijuana was a contributing factor in over 119,000 emergency department (ED) visits in the United States, with about 15 percent of the patients between the ages of 12 and 17, and almost two-thirds male.
  • DAWN Drug Abuse Warning Network
  • ADAM Advanced Driver Assistance Program
  • Cannabis use is world-wide public health issue. According to the
  • Marijuana is the most widely used illicit drug in the world. It has been estimated that one in 11 cannabis users will become dependent (Anthony et al., Clin Psychopharmacol, 1994); rates of cannabis dependence in several countries (e.g., Australia, USA, South Africa) have increased substantially over the past decade (Bhana et al., S Afr Med J, 2002; SAMHSA, 2003), as well as the number of individuals seeking treatment (Stephens et al., Clin Psychol, 1993; Treatment Episode Data Set, 2002).
  • a number of pathologic states affect the human cannabinoid system, including Alzheimer's disease, schizophrenia, depression, alcoholism, Parkinson's disease, stroke, premature labor, endotoxic shock, hepatic cirrhosis, atherosclerosis, cancer, bone implantation, glaucoma, emesis, and pruritus of various etiology (Pertwee, AAPS Journal, 2005; Mackie Annu Rev Pharmacol Tox, 2006).
  • CB 2 cannabinoid agonists have decided to focus on CB 2 cannabinoid agonists since it is assumed that CB 2 receptors are found mainly outside of the brain, in immune cells and that CBj receptors are found throughout the body, but primarily in the brain.
  • cannabinoid agonist An important drawback with the use of cannabinoid agonist is the risk of drug addiction, drug diversion and drug abuse.
  • unsuspecting clinicians may prescribe a cannabinoid agonist to patients who have an underlying, undisclosed addiction disorder.
  • unsuspecting clinicians may prescribe a cannabinoid agonist to patients with a medical condition amenable to treatment or prevention with a cannabinoid agonist who in turn divert a portion of their prescribed dose to other individuals for nonmedical use.
  • psychoactive drugs There have also been documented cases of inappropriate prescribing or dispensing of psychoactive drugs by physicians and pharmacists, with its eventual diversion into the non-medical marketplace. Additionally, experience with other classes of psychoactive, abusable drugs suggest that non-medical supplies of pharmaceutical grade cannabinoid agonist will be available through prescription forgeries and break-ins into pharmacies.
  • compositions containing cannabinoid agonists will likely be used for non-medical purposes in a variety of settings: i) by patients with a medical condition amenable to treatment or prevention with a cannabinoid agonist who have developed an addiction disorder following initiation of the cannabinoid agonist; ii) by patients with an addiction disorder seeking cannabinoid agonists for their euphoriant properties and iii) by recreational drug users who may use cannabinoid agonists from time to time (“chippers”) for pleasure seeking effects, analogous to the intermittent use of marijuana by many users.
  • Cannabinoids hold substantial promise for the prevention and treatment of a wide variety of medical conditions, including multiple sclerosis, amyotrophic lateral sclerosis, encephalitis, Alzheimer's disease, Parkinson's disease, Huntington's disease, obesity, feeding, fasting, stress, schizophrenia, depression, alcoholism, stroke, premature labor, endotoxic shock, hepatic cirrhosis, atherosclerosis, pruritus of various etiology and cancer.
  • cannabinoid agonists are going to be widely used for a variety of pathologic states.
  • cannabinoid agonists In view of its anticipated widespread use, cannabinoid agonists also have the potential to create a major epidemic of drug abuse involving an entirely new pharmacologic class of agents. [0023] Epidemiologic data suggest that pharmaceutical cannabinoid agonists are also likely to be co-abused with alcohol.
  • Methylphenidate is a CNS stimulant with effects similar to, but more potent than, caffeine and less potent than . amphetamines. It has a paradoxical effect of calming and "focusing" efforts in individuals with ADHD, particularly children. Methylphenidate is an important therapeutic option for adults as well as children with ADHD. Due to its stimulant properties, however, in recent years there have been reports of methylphenidate abuse by people for whom it is not prescribed. It is abused for its stimulant effects: appetite suppression, wakefulness, increased focus/attentiveness, and euphoria.
  • Methylphenidate has been shown to serve as a positive reinforcer in monkeys (Johanson and Schuster, 1975, J Pharmacol Exp Ther; Wilson et al., 1971, Psycopharmacol), and has established potential for abuse (Wang et al., 1997, Eur Addict Res). In one study, methylphenidate consistently maintained a cocaine response in an animal model of cocaine addiction predictive of abuse potential in humans (Wee and Wolverton, Drug and Alcohol Dependence, 2004).
  • Methylphenidate is frequently the subject of theft from pharmacies and drug wholesalers. Organized drug-trafficking groups utilize various schemes to obtain methylphenidate for sale on the illicit market. There is also considerable diversion of methylphenidate. In one study, children and adolescents who had been prescribed methylphenidate were surveyed. The study found that approximately 20% of patients had been approached to sell, provide at no cost or trade their medicine at least once in the previous five 5 years (Musser et al., 1998).
  • benzodiazepines Another class of drugs that have significant potential for abuse, misuse and diversion are the benzodiazepines.
  • benzodiazepines served as effective reinforcers for rhesus monkeys and provide further evidence that benzodiazepines share significant characteristics with other drug reinforcers. (Gomez et al, 2002; Gomez et al., 2002; Stewart et al., 1994).
  • benzodiazepines prescription rates for benzodiazepines in general (Pincus et al., 1998) and for benzodiazepine anxiolytics among youths 20 years and adolescents (Zito et al., 2003).
  • benzodiazepines are safe and effective when used as intended, their increased use has raised public health concerns because of they also carry a high propensity for abuse.
  • the nonmedical use of benzodiazepines among adolescents and young adults has been on the increase (Gledhill-Hoyt et al., 2000; Johnston et al., 2003; Mohler-Kuo et al., 2003; SAMHSA, 2002).
  • DAWN Drug Abuse Warning Network
  • abusable drugs e.g., cannabinoid agonists, benzodiazepine agonists, and stimulant drugs
  • abusable drugs e.g., cannabinoid agonists, benzodiazepine agonists, and stimulant drugs
  • cannabinoid agonists e.g., cannabinoid agonists, benzodiazepine agonists, and stimulant drugs
  • this may be achieved by the use of extended release dosage forms that provide sustained therapeutic plasma concentrations.
  • extended release oral dosage forms are designed to maintain effective plasma levels throughout an 8, 12 or 24-hour dosing interval.
  • extended release formulations have now become the standard of care. Use of extended release dosage forms can result in fewer interruptions in sleep, reduced dependence on caregivers, improved compliance, enhanced quality of life outcomes, and increased control over the management of their disease.
  • extended release formulations can be important in reducing the need for supervised dosing in school or at home.
  • extended release formulations can provide more constant plasma concentrations and clinical effects, less frequent peak to trough fluctuations and fewer side effects, compared with short acting drugs.
  • Pharmaceutical dosage forms containing abusable drugs may be ingested whole, crushed and ingested, crushed or vaporized and snorted or injected intravenously after attempted extraction of the active pharmaceutical ingredient.
  • ADHD has revolutionized the management of many chronic conditions.
  • the widespread use of extended release dosage form of abusable drugs has also associated with its diversion into the non-medical supply for use both by addicts and recreational drug users.
  • Intentional or inadvertent tampering from extended release formulations will rapidly deliver a massive dose and produce profound a variety of serious and life threatening side effects, including, depending on the pharmacology of the abusahle drug, respiratory depression and stimulation, cardiac arrhythmias, sedation and hypersomnia, seizures, hypertension and hypertensive crises, cognitive and perceptual impairment, cardiovascular collapse, coma and death.
  • Addicts and recreational drug may use extended release abusable drugs by the parenteral, intranasal or oral route.
  • Dosage form abuse can involve physical, mechanical, thermal or chemical tampering of the dosage form (e.g., crushing, melting, solvent extraction and filtration)
  • Palladone® capsules were withdrawn from the market in both countries due to dose-dumping when co-ingested with alcohol.
  • patients consuming 240 mL of 40% ethanol had a 6-fold mean increase in peak plasma hydromorphone concentration compared with co-ingestion of Palladone® capsules with water.
  • One subject experienced a 16-fold increase when the drug was ingested with 40% alcohol compared with water.
  • Patients consuming 240 mL of 20% ethanol had a 2-fold mean increase in peak plasma hydromorphone concentration.
  • One subject in this group experienced a 6-fold increase when the drug was ingested with 20% alcohol compared with water.
  • Extended release formulations have become highly preferable and in some cases, the standard of care for the management of a wide variety of conditions, particularly chronic conditions. Additionally, extended release formulations can make otherwise non-viable pharmaceutical agents (e.g., due to an exceedingly short duration of effect) into viable formulations with clinical and commercial potential.
  • Nonlimiting examples include abusable drugs for the management of:
  • ADHD which can provide continuous suppression of hyperactivity and inattention with morning parental or self-administration, without the need for supplemental (e.g., noon-time) dosing may a potentially non-compliant child or adolescent or the need for administration by a teacher or school nurse; and (ii) sleep, where sustained delivery may provide optimal control of insomnia with reduced sleep interruptions and improved polysomnography outcomes.
  • Toxicity from abusable drugs can result from unintentional or intentional tampering of abusable drugs. It is not uncommon for patients who have difficulty swallowing, to crush the contents of tablets or open a capsule, and swallow the contents with liquids or on soft food. In the case of most immediate release dosage forms of abusable drugs, this generally produces no significant harm, with marginally higher peak concentrations (C max ) and time to peak concentrations (t max ). However, in the case of extended release dosage forms of abusable drugs, crushing the oral solid dosage form destroys the controlled-release mechanism and results in a rapid surge of drug into the bloodstream, with the entire 8, 12 or 24-hour drug supply released immediately with toxic effects, or pleasurable effects in the case of a drug abuser. For this reason, all extended release dosage forms available for sale in the United States carry a warning to the prescriber and patient not to crush or tamper with the oral solid dosage form.
  • An ideal formulation will provide a extended release pharmacokinetic profile suitable for every 8, 12 or 24 hour release and will be resist or frustrate attempts at crushing at room temperature and upon freezing, melting to allow for filtration and/or aspiration into a syringe and extraction with recreational solvents, all without doing harm to pain patients or patients with a substance abuse disorder, through the use of aversive agents or pharmacologic antagonists.
  • compositions containing abusable drugs have been used for non-medical purposes in a variety of settings: i) by patients with a disorder requiring treatment with an abusable drug who have developed an addiction disorder following initiation of therapy; ii) by patients with said disorder who had a pre-existing addiction disorder; iii) by patients with an addiction disorder seeking abusable drugs for their reinforcing, rewarding, euphoriant or other mood altering properties.
  • Non-medical users of abusable drugs are either recreational drug users who may use such agents episodically, or individuals with an addiction disorder who may require frequent maintenance doses.
  • Abusable drugs may be ingested whole, crushed and ingested, crushed or vaporized and snorted or injected intravenously after attempted extraction of the active pharmaceutical ingredient.
  • One mode of abuse can involve the extraction of the abusable drugs component from the dosage form by first mixing the tablet or capsule with a suitable solvent (e.g., water or alcohol), and then filtering and/or extracting the abusable drugs component from the mixture for intravenous injection.
  • a suitable solvent e.g., water or alcohol
  • Another mode of abuse of extended release abusable drugs can involve dissolving the drug in water, alcohol or another "recreational solvent" to hasten its release and to ingest the contents orally, in order to provide high peak concentrations and maximum euphoriant effects.
  • Scheduling of abusable drugs has also had the unintentional side-effect of causing physicians, fearful of being accused of permitting or even promoting drug abuse and drug overuse, to prescribe suboptimal doses of abusable drugs to patients in need of them, and to prescribe less effective drugs to patients that are not similarly scheduled.
  • abusable drugs An additional issue with extended release forms of drugs, including abusable drugs is the interaction of the drug, even in an untampered form, r when consumed with alcohol. Under such conditions, a number of drugs have demonstrated an in vitro and in vivo propensity for significant dose dumping when they are co-ingested for medical purposes at prescribed doses with alcohol, increasing the potential for drug toxicity and further exacerbating the intensity of the (abusable) drug-alcohol pharmacodynamic interaction.
  • the present invention also involves oral pharmaceutical compositions of abusable drugs, including in extended release form and methods of use thereof which provide reduced variability in rate and extent of absorption when taken with food, compared with the fasted state.
  • the U.S. prescribing information for OpanaTM ER states "two studies examined the effect of food on the bioavailability of single doses of 20 and 40 mg of OPANA ER in healthy volunteers. In both studies, after the administration of OPANA ER, the Cmax was increased by approximately 50% in fed subjects compared to fasted subjects. A similar increase in Cmax was also observed with oxymorphone solution.”
  • the U.S. prescribing information for Aderall XR TM (amphetamine and dextroamphetamine ER) states "Food does not affect the extent of absorption of d-amphetamine and 1- amphetamine, but prolongs T max by 2.5 hours (from 5.2 hrs at fasted state to 7.7 hrs after a high-fat meal) for d-amphetamine and 2.1 hours (from 5.6 hrs at fasted state to 7.7 hrs after a high fat meal) for 1- amphetamine after administration of ADDERALL XR M 30 mg.”
  • the present invention is directed at pharmaceutical compositions of abusable drugs to provide abuse deterrence properties.
  • the present invention is directed at pharmaceutical compositions of abusable drugs to provide extended release properties.
  • the present invention is directed at pharmaceutical compositions of abusable drugs that provide simultaneous abuse deterrence properties and extended release properties.
  • the present invention is directed at pharmaceutical compositions of abusable drugs that provide simultaneous abuse deterrence properties and extended release properties using substantially the same ingredients to achieve abuse deterrence properties and extended release.
  • the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide abuse deterrence properties.
  • the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide extended release properties.
  • the present invention is directed at oral solid pharmaceutical compositions of abusable drugs that are in the form of a liquid, semisolid, oil or otherwise difficult to granulate.
  • the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide simultaneous abuse deterrence properties and extended release properties.
  • the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide simultaneous abuse deterrence properties and extended release properties using substantially the same ingredients to achieve abuse deterrence properties and extended release.
  • the present invention is directed at oral abusable drug pharmaceutical compositions and the use thereof for preventing or minimizing the risk of abusable drug toxicity from either intentional or unintentional tampering.
  • the present invention is directed at oral abusable drug pharmaceutical compositions and the use thereof for deterring abuse by drug addicts and/or recreational drug users.
  • the present invention is directed at oral abusable drug pharmaceutical compositions that provide extended release delivery of the drug and the use thereof for the treatment of pain, addiction disorders, spasticity, musculoskeletal disorders, ADHD, insomnia, excessive sleepiness, daytime sleepiness, sleep disorders, anxiety disorders, panic attacks, agoraphobia, obsessive-compulsive disorders, psychiatric disorders, neurologic disorders, excess weight, obesity and other medical maladies responsive to treatment with the abusable drugs.
  • abusable drugs are, without limitation amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, nicotine, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof, but specifically excluding all opioid agonists.
  • abusable drugs are: (i) substances which have medical applications for the prevention or treatment of diseases and disorders (i.e., "therapeutically active agents”); and (ii) substances which have the potential for being abused by drug abusers, recreational drug users and individuals with an addiction disorder for intermittent use, recreational use or chronic use, wherein the abusable drug is being abused for one or more of the following effects: mood alterations; euphoria, pleasure; a feeling of high; a feeling of drug liking; anxiolysis; mental stimulation; increased mental arousal; sedation; calmness; a state of relaxation; psychotomimesis; hallucinations; alterations in perception, cognition and mental focus; insomnia; hypersomnia; increased wakefulness or alertness; memory improvement; increased sexual gratification; increased sexual arousal; increased sexual desire and sexual anticipation; increased socialization; reduced social
  • abusable drugs of the present invention can be formulated with the substantially the same ingredients to deter abuse and minimize abusable drug toxicity on tampering while simultaneously providing an extended release pharmacokinetic profile suitable for every 4, 6, 8, 12 or 24 hour dosing.
  • abusable drugs of the present invention can be formulated with the substantially the same ingredients to deter abuse and minimize abusable drug toxicity on tampering while simultaneously providing an extended release pharmacokinetic profile suitable for every 4, 6, 8, 12 or 24 hour dosing, without the need to include an aversive agent or an antagonist for the abusable drug in the formulation.
  • abusable drug pharmaceutical compositions and methods of the present invention provide (i) abuse deterrence; (ii) extended release; and (iii) simultaneous abuse deterrence and extended release, prepared using one or more ADER compounds.
  • abusable drug pharmaceutical compositions and methods of the present invention provide simultaneous abuse deterrence and extended release, prepared using ADER, using substantially the same ingredients to effect abuse deterrence and extended release.
  • abusable drug pharmaceutical compositions and methods of the present invention provide (i) extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER.
  • abusable drug pharmaceutical compositions and methods of the present invention provide (i) extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER, using substantially the same ingredients to effect extended release and protection against ethanol induced dose dumping.
  • abusable drug pharmaceutical compositions and methods of the present invention provide (i) abuse deterrence and extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER.
  • abusable drug pharmaceutical compositions and methods of the present invention provide (i) abuse deterrence and extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER, using substantially the same ingredients to effect abuse deterrence and extended release, and protection against ethanol induced dose dumping.
  • the present invention is directed to a novel method for reducing the peak concentration (C ma ⁇ ) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to a novel method for reducing the early post-dose partial area under the plasma concentration time curve (e.g., AUC 0 . 2 , AUCo-4 and AUCo-6) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • AUC 0 . 2 , AUCo-4 and AUCo-6 the early post-dose partial area under the plasma concentration time curve
  • the present invention is directed to a novel method for reducing the early post-dose average plasma concentration time (Cave) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • Cave early post-dose average plasma concentration time
  • the present invention is directed to a novel method for reducing the incidence of abusable drug toxicity upon tampering of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to a novel method for reducing the intensity of abusable drug toxicity upon tampering of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to a novel method for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including nausea, vomiting, somnolence, stupor, coma, respiratory depression, apnea, respiratory arrest, circulatory depression, bradycardia, hypotension, shock and skeletal muscle flaccidity, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to a novel method for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including tachycardia, mood alteration, euphoria, CNS stimulation, agitation, increased sweating, psychotomimetic effects, hallucinations, perception alterations, cognitive alterations, reinforcing effects and pleasurable effects said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to a novel method for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including "high”, “liking”, pleasurable, euphoric, alertness, wakefulness, calming, anxiolytic, auditory and visual perceptual alterations, relaxing, analgesic and rewarding effects.
  • the present invention is directed to novel pharmaceutical compositions for use in reducing the peak concentration (Cmax) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to novel pharmaceutical compositions for reducing the early post-dose partial area under the plasma concentration time curve (e.g., AUCo-2 5 AUCo-4 and AUC 0-6 ) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • novel pharmaceutical compositions for reducing the early post-dose partial area under the plasma concentration time curve e.g., AUCo-2 5 AUCo-4 and AUC 0-6
  • said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to novel pharmaceutical compositions for reducing the early post-dose average plasma concentration time (Cave) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to novel pharmaceutical compositions for reducing the incidence of abusable drug toxicity, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to novel pharmaceutical compositions for reducing the intensity of abusable drug toxicity, said method comprising administering the abusable drug and a suitable amount of ADER.
  • compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides for abuse deterrence of the abusable drugs.
  • abusable drugs e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benz
  • compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides extended release of the abusable drug.
  • abusable drugs e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine
  • compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g:, glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides abuse deterrence and extended release of the abusable drug.
  • abusable drugs e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate,
  • compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides simultaneous abuse deterrence and extended release of the abusable drug.
  • abusable drugs e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate
  • compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides simultaneous abuse deterrence and extended release of the abusable drug using substantially the same ingredients.
  • abusable drugs e.g., amphetamine and amphetamine like CNS-stimulants,
  • the present invention is directed to novel pharmaceutical compositions for reducing the intensity or frequency of one or more symptoms, including nausea, vomiting, somnolence, stupor, coma, respiratory depression, apnea, respiratory arrest, circulatory depression, bradycardia, hypotension, shock and skeletal muscle flaccidity, said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to novel pharmaceutical compositions for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including tachycardia, mood alteration, euphoria, CNS stimulation, agitation, increased sweating, psychotomimetic effects, hallucinations, perception alterations, cognitive alterations, reinforcing effects and pleasurable effects said method comprising administering the abusable drug and a suitable amount of ADER.
  • the present invention is directed to a novel pharmaceutical compositions for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including “high”; “liking”; “pleasurable”; “euphoric”; “alertness”; “wakefulness”; “calming”; “anxiolytic”; auditory and visual perceptual alterations; “relaxing” and “rewarding” effects.
  • the present invention is directed to a novel method and pharmaceutical compositions for preventing or minimizing excessive peak concentrations (dose dumping) of therapeutic doses of extended release abusable drugs used for medical purposes, when they are co- ingested with alcohol.
  • the present invention is directed to a novel method and pharmaceutical compositions for reducing the solvent extraction efficiency of the dosage form upon tampering.
  • the present invention is directed to a novel method and pharmaceutical compositions for reducing the filtration efficiency of the dosage form upon tampering.
  • the present invention is directed to a novel method and pharmaceutical compositions for preventing the surreptitious adulteration of beverages.
  • the present invention is directed .
  • pharmaceutical compositions which include one or more abusable drugs alone or in combination with other therapeutic agents, one or more ADER agents specified herein, and optionally one or more excipients (e.g., glidants, lubricants, disintegrants, etc) and inert carriers, said composition resisting, deterring, discouraging or preventing crushing, shearing, grinding, chewing, dissolving, melting, needle aspiration, inhalation, insufflation, solvent extraction and filtration of the abusable drug.
  • excipients e.g., glidants, lubricants, disintegrants, etc
  • compositions of the present invention provide a more extended release pharmacokinetic profile compared with formulations devoid of ADER.
  • compositions and methods of the present invention can form a viscous substance upon contact with a solvent such that the abusable drug cannot be easily drawn into a syringe; crushed and powdered to facilitate or enhance nasal delivery (snorting or nasal insufflation), inhalation or rapid oral delivery of a larger than medically intended delivery of the abusable drug; extracted with solvents and filtered.
  • the pharmaceutical composition resists the rapid release of all or substantially all of the abusable drug content of the unit dose upon tampering. In another preferred embodiment of the invention, the pharmaceutical composition resists the rapid release of a portion of the abusable drug content of the unit dose upon tampering. In yet another preferred embodiment of the invention, upon tampering, the abusable drug formulated with ADER resists the release of the abusable drug to a greater extent than when formulated without ADER.
  • the pharmaceutical composition resists the rapid release of all or substantially all of the abusable drug content of the unit dose upon co-administering with alcohol. In another preferred embodiment of the invention, the pharmaceutical composition resists the rapid release of a portion of the abusable drug content of the unit dose upon coadministering with alcohol. In yet another preferred embodiment of the invention, upon co-administering with alcohol, the abusable drug formulated with ADER resists the release of the abusable drug to a greater extent than when formulated without ADER.
  • the therapeutic pharmaceutical composition can be filled in a hard gelatin capsule without banding. In some preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be filled in a hard gelatin capsule with security banding. Li another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be filled in a soft shell capsules. In another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be prepared as a solid dispersion for direct compression into tablets, for extrusion and pelletization, followed by compression into a tablet or filling into a capsule. In another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be compressed into tablets. In another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be prepared into multiparticulate matrices followed by tabletting or capsule filling.
  • the present invention is directed at oral pharmaceutical compositions of abusable drugs or their pharmaceutically acceptable salts or mixtures thereof.
  • the present invention also relates to oral abusable drug pharmaceutical compositions and methods for the prevention and treatment of pain, musculoskeletal disorders, spasticity, ADHD, sleep disorders, anxiety, obsessive-compulsive disorders, psychiatric disorders, neurologic disorders, obesity and other medical conditions amenable to treatment with the abusable drug.
  • bioavailable oral formulations of abusable drugs suitable for up to once-daily (e.g
  • bioavailable oral extended release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with food, compared with the fasted state.
  • bioavailable oral extended release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with alcohol, compared to an alcohol-free state.
  • bioavailable formulations for oral administration suitable for up to once-a-day administration (e.g., Q4H, Q6H, Q8H, Ql 2H, and Q24H).
  • bioavailable formulations for oral administration suitable for up to once-a-day administration which provide an early onset and sustained duration of therapeutic effect.
  • the formulations of abusable drugs provide a therapeutic effect for up to about 30 minutes.
  • the formulations of abusable drugs provide a therapeutic effect for up to about 1 hour, or up to about 2 hours, or up to about 4 hours, or up to about 6 hours, or up to about 8 hours, or up to about 10 hours, or up to about 12 hours, or up to about 16 hours, or up to about 18 hours, or up to about 24 hours or up to about 36 hours, or up to about 48 hours.
  • tampering e.g., mechanical, thermal, chemical or physical tampering
  • the invention provides a method and formulations of oral abusable drugs for the prevention and treatment of pain, said formulations suitable for use in acute pain, including acute postsurgical pain.
  • the invention provides a method and formulations of oral abusable drugs for the prevention and treatment of chronic pain, cancer pain, neuropathic pain, somatic pain, visceral pain, idiopathic pain and breakthrough pain of various etiologies, including cancer, chronic pain and neuropathic pain.
  • sleep disorders e.e., insomnia, hypersomnia, narcolepsy, excessive daytime sleepiness
  • anxiety disorders e.g., generalized anxiety, agoraphobia, panic attacks
  • obsessive compulsive disorder e.g., through nicotine substitution or nicotine maintenance therapy
  • psychiatric disorders e.g., through nicotine substitution or nicotine maintenance therapy
  • addiction disorders e.g., through nicotine substitution or nicotine maintenance therapy
  • the oral dosage form of the present invention comprises a matrix which includes ADER and an abusable drug or a pharmaceutically acceptable salt thereof.
  • the matrix is compressed into a tablet and may be optionally overcoated with a coating that in addition to the sustained release material of the matrix may control the release of the abusable drug or pharmaceutically acceptable salt thereof from the formulation, such that blood levels of active ingredient are maintained within the therapeutic range over an extended period of time.
  • the matrix is encapsulated.
  • the sustained release oral dosage form of the present invention comprises ADER and a plurality of pharmaceutically acceptable sustained release matrices comprising an abusable drug or a pharmaceutically acceptable salt thereof, the dosage form maintaining the plasma levels of abusable drug within the therapeutic range over an extended period of time when administered to patients.
  • the abusable drugs are in a matrix that is in the form of pellets or beads.
  • the dosage form of the invention comprises a compressed tablet, compressed capsule or uncompressed capsule. In other embodiments, the dosage form comprises a liquid fill capsule.
  • the dosage form of the invention comprises an oral formulation (e.g., tablet or capsule) which is coated to prevent substantial direct contact of abusable drug with oral cavity (e.g. tongue, oral mucosa), oropharyngeal mucosal surface, esophagus or stomach.
  • abusable drug e.g., tablet or capsule
  • oral cavity e.g. tongue, oral mucosa
  • oropharyngeal mucosal surface e.g. tongue, oral mucosa
  • the dosage form of the invention comprises an oral formulation which is coated with a film or polymer.
  • the dosage form of the invention comprises abusable drugs in an enteric coating.
  • the dosage form of the invention comprises abusable drugs formulated with pharmaceutical excipients and auxiliary agents known in the art, such that the abusable drug is released after a approximately specific amount of time, or at an approximately specific anatomic location in the gastrointestinal tract, or when the dosage form is in contact with specific gastrointestinal conditions (e.g., pH range, osmolality, electrolyte content, food content.).
  • abusable drugs formulated with pharmaceutical excipients and auxiliary agents known in the art, such that the abusable drug is released after a approximately specific amount of time, or at an approximately specific anatomic location in the gastrointestinal tract, or when the dosage form is in contact with specific gastrointestinal conditions (e.g., pH range, osmolality, electrolyte content, food content.).
  • the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined under fed conditions. In other preferred embodiments, the in vivo pharmacokinetic parameters are derived or determined under fasted conditions.
  • compositions and methods of the present invention specifically exclude opioids, opioid agonists and opioid analgesics.
  • compositions and methods of the present invention exclude the prevention or treatment of pain.
  • compositions and methods of the present invention involve abusable drugs and ADER, said compositions and methods excluding all drugs for the management of pain.
  • abusable drugs specifically excludes all opioid agonists.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS -stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; and (iv) cannabinoid agonists.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; and (iv) cannabinoid agonists.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; and (iv) cannabinoid agonists.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances which are scheduled drugs under the United States Controlled Substances Act of 1970, as amended.
  • Controlled Substances Act of 1970 and "United States Controlled Substances Act of 1970” specifically excludes opioid agonists.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended); (vi) drugs listed under the United States Psychotropic Substances Act of 1978, as amended.
  • abusable drugs are limited to: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cann
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended).
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended); (vi) drugs listed under the United States Psychotropic Substances Act of 1978, as amended.
  • abusable drugs include: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof.
  • the abusable drugs include amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics, nicotine, nicotine analogs, tobacco extract, and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof.
  • the abusable drugs include amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, methylphenidate, alkylxanthines, anorectics, psychosti
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to amphetamine and amphetamine like CNS-stimulants.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to methylphenidate, methylphenidate prodrugs and methylphenidate esters.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to benzodiazepine agonists
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to barbiturates.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to cannabinoid agonists.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to prodrugs of abusable drugs.
  • compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to nicotine.
  • the dosage form is a capsule, said capsule rendered tamper-resistant with a security banding seal between the capsule parts.
  • the dosage form is a capsule, said capsule rendered liquid tight or leak resistant with a banding seal between the capsule parts.
  • the invention relates to a method and pharmaceutical compositions for prevention or treatment of (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive- compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug; comprising oral administration of a dosage form • containing an abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER.
  • the dosage form provides an oral pharmaceutical composition for the prevention and treatment of (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug; comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form: (a) abuse deterrent; and/or (b) extended release; and/or (c) resistant to significant alcohol dose-dumping; and/or (d) resistant
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form abuse deterrent, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency, hi other preferred embodiments, the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug .or a mixture thereof and ADER material to render said dosage form extended release; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency, hi other preferred embodiments, the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form abuse deterrent and extended release, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency.
  • the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form resistant to alcohol dose dumping, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency.
  • the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form resistant to significant fluctuations in bioavailability when given under fed and fasted conditions, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency.
  • the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
  • the invention comprises an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug, or a mixture thereof and ADER to render said dosage form suitable for three times a day administration (TID) or about every eight hours administration (Q8H).
  • the TED or Q8H oral pharmaceutical composition of an abusable drug provides a therapeutic effect for about 8 hours.
  • the TDD or Q8H oral pharmaceutical composition of an abusable drug provides a C ma ⁇ of abusable drugs at about 1 to about 6 hours.
  • the TID or Q8H oral pharmaceutical composition of abusable drugs provide a C min of abusable drugs at about 6 to 10 hours.
  • the TID or Q8H oral pharmaceutical composition of abusable drugs provide a mean abusable drugs Cs/Cmax ratio of 0.25 to about 0.95.
  • the TDD or Q8H oral pharmaceutical composition of abusable drugs provide a percent fluctuation of less than 400%.
  • the TED or Q8H oral pharmaceutical composition of an abusable drug provides a W 50 of 1.5 to about 6.5 hours.
  • the TDD or Q8H oral pharmaceutical composition of an abusable drug provides an HVD of 2 to about 7 hours.
  • the TDD or Q8H oral pharmaceutical composition of an abusable drug provides an HVD of about 2 to about 7 hours.
  • the TDD or Q8H oral pharmaceutical composition of an abusable drug an AI of not more that 4.0.
  • the invention comprises an oral pharmaceutical composition comprising therapeutically effective amounts of abusable drug or pharmaceutically acceptable salts thereof, or mixtures thereof and ADER; said dosage from providing a C max of abusable drug occurring from a mean of about 0.25 to about 30 hours.
  • the dosage form provides a C ma ⁇ of abusable drug occurring from a mean of about 0.5 to about 30 hours, or from a mean of about 1 to about 30 hours, or about 1 to about 26 hours, or about 1 to about 24 hours, or about 1 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours, or about 1 to about 14 hours, or about 1 to about 12 hours, or about 1 to about 10 hours, or about 1 to about 8 hours, or about 1 to about 6 hours, or about 1 to about 4 hours, or about 1 to about 3 hours, or about 2 to about 30 hours, or about 4 to about 30 hours, or about 4 to about 24 hours, or about 6 to about 24 hours, or about 8 to about 24 hours, or about 10 to about 20 hours, or about 12 to about 24 hours, or about 18 to about 24 hours, or about 2 to about 12 hours, or about 3 to about 12 hours, or about 3 to about 8 hours, or about 4 to about 10 hours, or about 4 to about 12 hours, or about 4 to about 9
  • the invention comprises an oral pharmaceutical composition comprising therapeutically effective amounts of abusable drug or pharmaceutically acceptable salts thereof, or mixtures thereof and ADER; said dosage from providing a C m j n of abusable drug occurring from a mean of about 0.5 to about 28 hours, or about 1 to about 28 hours, or about 1 to 24 hours, or about 1 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours, or about 1 to about 12 hours, or about 1 to 10 hours, or about 1 to about 8 hours, or about 1 to about 6 hours, or about I to about 4 hours, about 2 to about 24 hours, or about 3 to 24 hours, or about 4 to about 24 hours, or about 6 to about 24 hours, or about 8 to about 24 hours, about 2 to about 12 hours, or about 3 to 10 hours, or about 3 to about 8 hours, or about 4 to about 8 hours, or about 6 to about 10 hours.
  • the invention comprises an oral pharmaceutical composition comprising therapeutically effective amounts of abusable drug or pharmaceutically acceptable salts thereof, or mixtures thereof and ADER; said dosage form providing a systemic exposure as assessed by the mean abusable drug area under the plasma concentration time curve (AUCo-O after first administration which is at least about 40% of the area under the plasma drug concentration-time curve from time zero to infinity (AUCo-).
  • AUCo-O mean abusable drug area under the plasma concentration time curve
  • AUCo-O after first administration which is at least about 40% of the area under the plasma drug concentration-time curve from time zero to infinity
  • the dosage from provides an AUCo- t which is at least about 45%, or which is at least about 50%, or which is at least about 55%, or at least about 60%, or which is at least about 65%, or at least about 70%, or which is at least about 75%, or at least about 80%, or at least about 85%, or at least about 88%, or at least about 90%, or at least about 92%, or at least about 94%, or at least about 96% or at least about 98% of the AUC 0- .,.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of an abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form providing at least 80% of the steady state therapeutic concentration of abusable drug after administration of ⁇ three doses at their intended dosing frequency.
  • said dosage form provides at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 85%, or at least about 90%, or at least about 92%, or at least about 95%, or at least about 97%, or at least about 99% of the steady state therapeutic concentration of abusable drug after administration of ⁇ three doses at their intended dosing frequency.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form providing at least 80% of the steady state therapeutic concentration of abusable drug after administration of ⁇ two doses at their intended dosing frequency.
  • said dosage form provides at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 85%, or at least about 90%, or at least about 92%, or at least about 95%, or at least about 97%, or at least about 99% of the steady state therapeutic concentration of abusable drug after administration of ⁇ two doses at their intended dosing frequency.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form providing at least 80% of the steady state therapeutic concentration of abusable drug after administration of one dose at their intended dosing frequency.
  • said dosage form provides at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 85%, or at least about 90%, or at least about 92%, or at least about 95%, or at least about 97%, or at least about 99% of the steady state therapeutic concentration of abusable drug after administration of one dose at their intended dosing frequency.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing a C m ⁇ n /C max ratio of abusable drug of 0.1 to about 1.0.
  • the dosage form provides a C m i n /C max ratio of abusable drug of about 0.1 to about 0.9, or about 0.1 to about 0.8, or about 0.1 to about 0.7, or about 0.1 to about 0.6, or about 0.1 to about 0.5, or about 0.1 to about 0.4, or about 0.1 to about 0.3, or about 0.2 to about 1.0, or about 0.25 to about 1.0, or about 0.4 to about 1.0, or about 0.5 to about 0.9, or about 0.5 to about 0.85, or about 0.5 to about 0.8, or about 0.5 to about 0.75, or about 0.5 to about 1.0, or about 0.65 to about 1.0, or about 0.75 to about 1.0, or about 0.2 to about 0.9, or about 0.3 to about 0.95, or about 0.3 to about 0.85,or about 0.3 to about 0.8, or about 03 to about 0.75, or about 0.3 to about 0.7, or about 0.3 to about 0.6, or about 0.4 to about 0.9, or
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing a percent fluctuation of abusable drug of less than 400%.
  • the dosage form provides a percent fluctuation of abusable drug of less than 350%, or less than 300%, or less than 250%, or less than 200%, or less than 150%, or less than 100%, or less than 75%, or less than 50%, or less than 25%.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing a W 50 of abusable drug of about 1 to about 6 hours for each 6 hour time period of intended dosing frequency and intended duration of action.
  • the dosage form provides a W 5 0 of abusable drug for each 6 hour time period of intended dosing frequency and intended duration of action of about 1 to about 5 hours, or about 1 to about 4 hours, or about 1 to about 3 hours, or about 1 to about 2 hours, or 2 to about 6 hours, or about 3 to about 6 hours, or about 4 to about 6 hours, or about 2 to about 4 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing an HVD of abusable drug of about 1.5 to about 6 hours for each 6 hour time period of intended dosing frequency and intended duration of action.
  • the dosage form provides a HVD of abusable drug for each 6 hour time period of intended dosing frequency and intended duration of action of about 1.5 to about 5 hours, or about 1.5 to about 4 hours, or about 1.5 to about 3 hours, or about 1.5 to about 2 hours, or 2 to about 6 hours, or about 3 to about 6 hours, or about 4 to about 6 hours, or about 2 to about 4 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing an AI of abusable drug of not more than 3.0.
  • the dosage form provides an AI of abusable drug of not more than about 2.5, or not more than about 2, or not more than about 1.75, or not more than about 1.5, or not more than about 1.25, or not more than about 1, or not more than about 0.75, or not more than about 0.5, or not more than about 0.25.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form providing a C max of abusable drug at 2 to about 10 hours; and said dosage form providing a therapeutic effect for at least about 12 hours.
  • the dosage form provides a C max of abusable drug at about 2 to abo ⁇ t 8 hour or about 2 to about 6 hours, or about 2 to about 5 hours, or about 2 to about 7 hours, or about 2 to about 4.5 hours, or about 2 to about 4 hours, or 2 to about 3.5 hours, or about 2 to about 3 hours, or about 3 to about 10 hours, or about 3.5 to about 10 hours, or about 4 to about 10 hours, or about 4.5 to about 10 hours, or about 5 to about 10 hours, or 5 to about 10 hours, or about 6 to about 10 hours, or about 3 to about 8 hours, or about 3 to about 7 hours, or about 3 to about 6 hours, or about 4 to about 8 hours, or about 4 to about 6.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form providing a Ci2/Cma X ratio of abusable drug of 0.1 to about 1; and said dosage form providing a therapeutic effect for at least about 12 hours.
  • the dosage form provides a C)2/C max ratio of abusable drug of about 0.25 to about 0.9, or about 0.25 to about 0.8, or about 0.25 to about 0.75, or about 0.25 to about 0.6, or 0.25 to about 0.5, or about 0.25 to about 0.4, or about 0.25 to about 0.35, or about 0.3 to about 0.95, or about 0.4 to about 0.95, or about 0.5 to about 0.95, or about 0.65 to about 0.95, or about 0.75 to about 0.95, or about 0.3 to about 0.8, or about 0.4 to about 0.75, or about 0.5 to about 0.75, or about 0.1 to about 0.9, or about 0.1 to about 0.8, or about 0.1 to about 0.7, or about 0.1 to about 0.6, or about 0.1 to about 0.5, or about 0.1 to about 0.4, or about 0.1 to about 0.3, or about 0.2 to about 1.0, or about 0.25 to about 1.0, or about 0.4 to about 1.0, or about 0.5 to about 0.5 to about 0.9
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form providing a percent fluctuation of abusable drug of less than 400%; and said dosage form providing a therapeutic effect for at least about 12 hours.
  • the dosage form provides a percent fluctuation of abusable drug of less than about 375%, or less than about 350%, or less than about 325%, or less than about 300%, or less than about 275%, or less than about 250%, or less than about 225%, or less than about 200%, or less than about 175%, or less than about 150%, or less than about 125%, or less than about 100%, or less than about 75%; or less than about 50%, or less than about 25%.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form after administration to a human patient, providing a W 50 of abusable drug of 2 to about 11 hours; and said dosage form providing a therapeutic effect for at least about 12 hours.
  • the dosage form provides a W 50 of abusable drug of about 2 to about 10 hours, or about 2 to about 9 hours, or about 2 to about 9 hours, or about 2 to about 8 hours, or 2 to about 7 hours, or.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form after administration to a human patient, providing a HVD of abusable drug of 1.5 to about 10 hours; and said dosage form providing a therapeutic effect for at least about 12 hours.
  • the dosage form provides an HVD of abusable drug of about 1.5 to about 9 hours, or about 1.5 to 8 hours, or about 1.5 to about 7 hours, or about 1.5 to 6 hours, or about 1.5 to about 5 hours, or about 1.5 to about 4 hours, or about 2 to about 10 hours, or about 3 to 10 hours, or about 4 to about 10 hours, or about 5 to 10 hours, or about 6 to about 10 hours, or about 8 to 10 hours, about 3 to about 8 hours, or about 4 to 8 hours, or about 5 to about 7 hours, or about 3 to .6 hours, or about 3 to about 8 hours, or about 5 to about 8 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form after administration to a human patient, providing an AI of abusable drug of not more that 4.0; and said dosage form providing a therapeutic effect for at least about 12 hours.
  • the dosage form provides an AI of abusable drug of not more than about 3.75, or not more than about 3.5, or not more than about 3.25, or not more than about 3, or not more than about 2.75, or not more than about 2.5, or not more than about 2, or not more than about 1.5, not more than about 1.25, or not more than about 1, or not more than about 0.75.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours.
  • the dosage form provides said an in-vitro release rate of from 0% to about 40% at 1 hour, from about 5% to about 55% at 2 hours, from about 10% to about 60% at 4 hours, from about 15% to about 70% at 6 hours, from about 25% to about 80% at 9 hours, and greater than about 50% at 12 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours; said dosage form providing a Cm a x from a mean of about 2 to about 10 hours after first administration or at steady state.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours-, and greater than about 65% at 12 hours; said dosage form providing a C m i n occurring from a mean of about 10 to about 14 hours after first administration or at steady state.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours; said dosage form providing a mean abusable drug AUCoVAUCo- ⁇ ratio after first administration of about 0.4, or about 0.5, or about 0.6, or about 0.7, or
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours; said in-vitro release rate being substantially independent of pH in that a difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH, when
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a C max of abusable drug at about 3 to about 20 hours; and said dosage form providing a therapeutic effect for at least about 24 hours.
  • the abusable drugs dosage forms provide a C max of abusable drug at about 3 to about 18 hours, or about 3 to about 15 hours, or about 3 to about 12 hours, or at about 3 to about 10 hours, or at about 3 to about 8 hours, or at about 3 to about 7 hours, or at about 3 to about 7 hours, or about 4 to about 20 hours, or about 5 to about 20 hours, or about 6 to about 20 hours, or at about 8 to about 20 hours, or at about 10 to about 20 hours, or at about 12 to about 20 hours, or at about 14 to about 20 hours, or about 18 to about 20 hours, or about 4 to about 18 hours, or about 4 to about 16 hours, or at about 4 to about 12 hours, or at about 4 to about 8 hours, or at about 4 to about 10 hours, or at about 3 to about 6 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a C m i n of abusable drug at about 20 to about 28 hours; and said abusable drugs dosage forms providing a therapeutic effect for at least about 24 hours.
  • the abusable drugs dosage forms provide a C m i n of abusable drug at about 20 to about 26 hours, or about 20 to about 27 hours, or about 20 to about 25 hours, or about 20 to about 24 hours, or about 20 to about 23 hours, or about 21 to about 28 hours, or about 22 to about 28 hours, or about 23 to about 28 hours, or about 23.5 to about 28 hours, or about 22 to 26 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage from providing a C ma ⁇ of abusable drug from about 0.25 hours to about 30 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage from providing a C m i n of abusable drug from about 0.5 hour to about 30 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a C 24 ZC m3x ratio of abusable drug of 0.1 to about 1; and said dosage form providing a therapeutic effect for at least about 24 hours.
  • the dosage form provides a C 2 4/C max ratio of abusable drug of about 0.25 to about 0.9, or about 0.25 to about 0.8, or about 0.25 to about 0.75, or about 0.25 to about 0.6, or 0.25 to about 0.5, or about 0.25 to about 0.4, or about 0.25 to about 0.35, or about 0.3 to about 0.95, or about 0.4 to about 0.95, or about 0.5 to about 0.95, or about 0.65 to about 0.95, or about " 0.75 to about 0.95, or about 0.3 to about 0.8, or about 0.4 to about 0.75, or about 0.5 to about 0.75, or about 0.1 to about 0.9, or about 0.1 to about 0.8, or about 0.1 to about 0.7, or about 0.1 to about 0.6, or about 0.1 to about 0.5, or about 0.1 to about 0.4, or about 0.1 to about 0.3, or about 0.2 to about 1.0, or about 0.25 to about 1.0, or about 0.4 to about 1.0, or about 0.5 to
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a percent fluctuation of abusable drug of less than 400%; and said dosage form providing a therapeutic effect for at least about 24 hours.
  • the dosage form provides a percent fluctuation of abusable drug of less than about 375%, or less than about 350%, or less than about 325%, or less than about 300%, or less than about 275%, or less than about 250%, or less than about 225%, or less than about 200%, or less than about 175%, or less than about 150%, or less than about 125%, or less than about 100%, or less than about 75%, or less than about 50%, or less than about 25%.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said abusable drugs dosage form after administration to a human patient, providing a W 5 0 of abusable drug of 4 to about 22 hours; and said dosage form providing a therapeutic effect for at least about 24 hours.
  • the abusable drugs dosage from provides a W 50 of abusable drug of about 4 to about 20 hours, or about 4 to about 19 hours, or about 4 to about 18 hours, or 4 to about 16 hours, or 4 to about 14 hours, or about 4 to about 12 hours, or about 4 to about 10 hours, or about 4 to about 8 hours, or about 6 to about 20 hours, or about 8 to about 20 hours, or about 10 to about 20 hours, or about 12 to about 20 hours, or 14 to about 20 hours, or about 6 to about 16 hours, or about 8 to about 16 hours, or about 8 to about 14 hours, or about 6 to about 12 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said abusable drugs dosage form after administration to a human patient, providing a HVD of abusable drug of 3 to about 20 hours; and said dosage form providing a therapeutic effect for at least about 24 hours.
  • the abusable drugs dosage from provides an HVD of abusable drug of about 3 to about 18 hours, or about 3 to 16 hours, or about 3 to about 14 hours, or about 3 to 12 hours, or about 3 to about 10 hours, or about 3 to about 8 hours, or about 4 to about 20 hours, or about 6 to 20 hours, or about 8 to about 20 hours, or about 10 to 20 hours, or about 12 to about 20 hours, or about 16 to 20 hours, about 6 to about 16 hours, or about 8 to 16 hours, or about 10 to about 14 hours, or about 6 to 12 hours, or about 6 to about 16 hours, or about 10 to about 16 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form after administration to a human patient, providing an AI of abusable drug of not more that 4.0; and said abusable drugs dosage form providing a therapeutic effect for at least about 24 hours, hi other preferred embodiments, the abusable drugs dosage from provides an AI of abusable drug of not more than about 3.75, or not more than about 3.5, or not more than about 3.25, or not more than about 3, or not more than about 2.75, or not more than about 2.5, or not more than about 2, or not more than about 1.5, not more than about 1.25, or not more than about 1, or not more than about 0.75.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said abusable drug dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 °C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said abusable drug dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said dosage form providing a C m3x from a mean of about 3 to about 20 hours after first administration or at steady state.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said dosage form providing a C m i n of abusable drug occurring from a mean of about 20 to about 28 hours after first administration or at steady state.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said dosage form providing a mean abusable drug AUCo-t/AUCo- ⁇ ratio after first administration of about 0.4, or about 0.5, or about 0.6, or about 0.7, or about 0.75, or
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said in-vitro release rate being substantially independent of pH in that a difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH, when measured in-vitr
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for extended release administration to a human patient; said dosage form after administration to a human patient providing a mean abusable drug C m a x occurring from a mean of about 0.25 to about 22 hours; said dosage form providing a mean abusable drug C m j n occurring from a mean of about 0.5 to about 28 hours; said dosage form providing a mean abusable drug HVD of about 1 to about 5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug W 50 of about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug AI of not more than 3.0; said dosage form providing a mean abusable drug percent
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for extended release administration to a human patient; said dosage form after administration to a human patient providing a mean abusable drug Cm 3x which is less than 65% of the C ma ⁇ of an equivalent dose of an oral immediate release abusable drug solution or suspension; and said dosage form maintaining a mean abusable drug plasma concentration within 50% of C ma ⁇ for about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for extended release administration to a human patient; said dosage form after administration to a human patient providing a mean abusable drug C m3x occurring from a mean of about 0.25 to about 22 hours; said dosage form providing a mean abusable drug C m i n occurring from a mean of about 0.5 to about 28 hours; said dosage form providing a mean abusable drug HVD of about 1 to about 5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug W 50 of about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug AI of not more than 3.0; said dosage form providing a mean abusable drug percent fluctu
  • said dosage form providing a mean time to 75% abusable drug C max of about 100% to about 2000% of the time to 75% mean C max of an oral immediate release abusable drug solution or suspension; said dosage form after administration to a human patient providing a mean abusable drug C max which is less than 65% of the C max of an equivalent dose of an oral immediate release abusable drug solution or suspension; and said dosage form maintaining a mean abusable drug plasma concentration within 50% of C max for about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action.
  • the abusable drugs dosage forms provide an in- vitro release of from 0% to about 50% by weight of the abusable drug or a pharmaceutically acceptable salt thereof from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) at 37 0 C.
  • SGF Simulated Gastric Fluid
  • said in-vitro release rate by weight of the abusable drug or a pharmaceutically acceptable salt thereof from said dosage form is from about 5% to about 45% , or about 10% to about 50%, or about 5% to about 60%, or about 5% to about 70%, or about 5% to about 80%, or about 5% to about 90%, or about 5% to about 100%, or about 10% to about 20%, or about 10% to about 35% , or about 10% to about 50%, or about 10% to about 60%, or about 10% to about 70%, or about 10% to about 80%, or about 10% to about 90%, or about 10% to about 100%, or about 20% to about 40% , or about 20% to about 50%, or about 20% to about 60%, or about 20% to about 70%, or about 20% to about 80%, or about 20% to about 90%, or about 20% to about 100%, or about 30% to about 50%, or about 30% to about 60%, or about 30% to about 70%, or about 30% to about 80%, or about 30% to about 90%, or about 40% to about 80%, or about 40% to about 90%,
  • the abusable drugs dosage form provides a C maX of abusable drug which is less than 65% of the Cmax of an equivalent dose of an oral immediate release abusable drug solution or suspension.
  • said dosage form provides a C max which is less than about 85%, or less than about 75%, or less than about 60%, or less than about 55%, or less than about 50%, or less than about 45%, or less than about 40%, or less than about 30%, or less than about 20% of the C max of an equivalent dose of an oral immediate release abusable drug solution or suspension.
  • the dosage form provides a time to
  • the dosage form provides a time to
  • the dosage from maintains a plasma abusable drug concentration within 50% of C max for about 1 to about 9 hours during a 12 hour dosing interval.
  • said dosage form maintains plasma abusable drug concentration within 50% of C max for about 2 to about 9 hours, or about 3 to about 9 hours, or about 4 to about 9 hours, or about 5 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 11 hours, or about 2 to about 11 hours, or about 3 to about 11 hours or about 4 to about 11 hours, or about 5 to about 11 hours, or about 6 to about 11 hours, or about 7 to about 11 hours, or about 8 to about 11 hours, or about 1 to about 10 hours, or about 2 to about 10 hours, or about 3 to about 10 hours or about 4 to about 10 hours, or about 5 to about 10 hours, or about 6 to about 10 hours, or about 7 to about 10 hours, or about 8 to about 10 hours, or about 1 to about 7 hours, or about 2 to about 7 hours, or about 3
  • the dosage from maintains a plasma abusable drug concentration within 30% of C max for about 1.5 to about 9 hours during a 12 hour dosing interval.
  • said dosage form maintains plasma abusable drug concentration within 30% of C max for about 2 to about 9 hours, or about 3 to about 9 hours, or about 4 to about 9 hours, or about 5 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 11 hours, or about 2 to about 11 hours, or about 3 to about 11 hours or about 4 to about 11 hours, or about 5 to about 11 hours, or about 6 to about 11 hours, or about 7 to about 11 hours, or about 8 to about 11 hours, or about 1 to about 10 hours, or about 2 to about 10 hours, or about 3 to about 10 hours or about 4 to about 10 hours, or about 5 to about 10 hours, or about 6 to about 10 hours, or about 7 to about 10 hours, or about 8 to about 10 hours, or about 1 to about 7 hours, or about 2 to about 7 hours, or about 3
  • the dosage from maintains a plasma abusable drug concentration within 65% of C ma ⁇ for about 1 to about.9 hours during a 12 hour dosing interval.
  • said dosage form maintains plasma abusable drug concentration within 65% of C max for about 2 to about 9 hours, or about 3 to about 9 hours, or about 4 to about 9 hours, or about 5 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 11 hours, or about 2 to about 11 hours; or about 3 to about 11 hours or about 4 to about 11 hours, or about 5 to about 11 hours, or about 6 to about 11 hours, or about 7 to about 11 hours, or about 8 to about 11 hours, or about 1 to about 10 hours, or about 2 to about 10 hours, or about 3 to about 10 hours or about 4 to about 10 hours, or about 5 to about 10 hours, or about 6 to about 10 hours, or about 7 to about 10 hours, or about 8 to about 10 hours, or about 1 to about 7 hours, or about 2 to about 7
  • the dosage from maintains a plasma abusable drug concentration within 55% of Cmax for about 3 to about 22 hours during a 24 hour dosing interval.
  • said dosage form maintains plasma abusable drug concentration within 50% of C m3x for about 1 to about 9 hours, or about 4 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 20 hours, or about 2 to about 20 hours, or about 3 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours or about 2 to about 18 hours, or about 2 to about 16 hours, or about 1 to about 14 hours, or about 1 to about 12 hours, or about 4 to about 16 hours, or about 4 to about 18 hours, or about 4 to about 20 hours, or about 3 to about 15 hours or about 6 to about 15 hours, or about 6 to about 12 hours, or about 6 to about 18 hours, or about 6 to about 20 hours, or about 5 to about 12 hours, or about 5 to about 14 hours, or about 3 to about 22 hours,
  • the dosage from maintains a plasma abusable drug concentration within 30% of C ma ⁇ for about 2 to about 22 hours during a 24 hour dosing interval.
  • said dosage form maintains plasma abusable drug concentration within 30% of C n ⁇ x for about 1 to about 9 hours, or about 4 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 20 hours, or about 2 to about 20 hours, or about 3 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours or about 2 to about 18 hours, or about 2 to about 16 hours, or about 1 to about.14 hours, or about 1 to about 12 hours, or about 4 to about 16 hours, or about 4 to about 18 hours, or about 4 to about 20 hours, or about 3 to about 15 hours or about 6 to about 15 hours, or about 6 to about 12 hours, or about 6 to about 18 hours, or about 6 to about 20 hours, or about 5 to about 12 hours, or about 5 to about 14 hours, or about 3
  • the dosage from maintains a plasma abusable drug concentration within 65% of C ma ⁇ for about 2 to about 22 hours during' a 24 hour dosing interval.
  • said dosage form maintains plasma abusable drug concentration within 65% of C m ax for about 1 to about 9 hours, or about 4 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 20 hours, or about 2 to about 20 hours, or about 3 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours or about 2 to about 18 hours, or about 2 to about 16 hours, or about 1 to about 14 hours, or about 1 to about 12 hours, or about 4 to about 16 hours, or about 4 to about 18 hours, or about 4 to about 20 hours, or about 3 to about 15 hours or about 6 to about 15 hours, or about 6 to about 12 hours, or about 6 to about 18 hours, or about 6 to about 20 hours, or about 5 to about 12 hours, or about 5 to about 14 hours, or about
  • the dosage form provides a T max of abusable drug at a time point 1 to 18 times later than the T m a X provided by an equivalent dose of an oral immediate release abusable drug solution or suspension, hi the dosage form provides a T max at a time point about 1 to 15 times late, or about of 1 to 10 times later, or about of 1 to 7 times later, or about of 1 to 4 times later, or about of 3 to 20 times later, or about of 3 to 10 times later, or about of 3 to 5 times later, or about 1.5 to 15 times later, or about of 1.5 to 10 times later, or about of 1.5 to 7 times later, or about of 1.5 to 3 times later, or about of 2 to 20 times later, or about of 2 to 10 times later, or about of 2 to 5 times later, or about of 2 to 3 times later, or about of 2.5 to 20 times later, or about of 2.5 to 8 times later, or about of 2.5 to 5 times later, or about of 2.5 to 4 times later, or about of 3 to 20 times later, or
  • the dosage form provides a mean in vivo extent of absorption of abusable drug from 0 to 4 hours which is at least 20% of the mean in vivo extent of absorption from to 0 to 12 hours, wherein the mean in vivo extent of absorption is the area under the plasma or serum abusable drug concentration time curve from the time of drug administration to the specified time point.
  • said in vivo extent of absorption from 0 to 4 hours is at least about 5%, or at least about 10%, or at least about 15%, or at least about 25%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, at least about 90%, or about 100% of the mean in vivo extent of absorption from to 0 to 12 hours.
  • the dosage form provides a mean in vivo extent of absorption of abusable drug from 0 to 8 hours which is at least 20% of the mean in vivo extent of absorption from to 0 to 24 hours, wherein the mean in vivo extent of absorption is the area under the plasma or serum abusable drug concentration time curve from the time of drug administration to the specified time point.
  • said in vivo extent of absorption from 0 to 8 hours is at least about 5%, or at least about 10%, or at least about 15%, or at least about 25%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, at least about 90%, or about 100% of the mean in vivo extent of absorption from to 0 to 24 hours.
  • the dosage form provides a mean in vivo extent of absorption of abusable drug from 0 to 12 hours which is at least 20% of the mean in vivo extent of absorption from to 0 to 24 hours, wherein the mean in vivo extent of absorption is the area under the plasma or serum abusable drug concentration time curve from the time of drug administration to the specified time point, hi other preferred embodiments, said in vivo extent of absorption from 0 to 12 hours is at least about 5%, or at least about 10%, or at least about 15%, or at least about 25%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, at least about 90%, or about 100% of the mean in vivo extent of absorption from to 0 to 24 hours.
  • the dosage form provides a mean in vivo extent of absorption of abusable drug over the dosing interval, AUCo-t (e.g., from 0 to 8 hours, or from 0 to 12 hours or from 0 to 24 hours) which is at least 40% of the mean in vivo extent of absorption from to 0 to infinity (AUCo- ⁇ ).
  • said AUCo-t is at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% of the mean in vivo extent of absorption from to 0 to infinity
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% to about 100% at 0.5 hours, and greater than about 60% at 1 hour.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% to about 40% at 1 hour, from about 5% to about 60% at 2 hours, from about 10% to about 75% at 4 hours, from about 20% to about 75% at 6 hours, from about 30% to about 80% at 9 hours, and greater than about 70% at 12 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 1% and about 45% at 1 hour, between about 5% and about 70% at 2 hours, between about 10% and about 90% at 4 hours, between about 20% and about 90% at 8 hours, greater than about 60% at 12 hours, greater than about 80% at 18 hours, and greater than about 85% at 24 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between(1.6 and 7.2 at 37 0 C of between 5% and about 60% at 1 hour, between about 12.5% and about 80% at 2 hours, between about 25% and about 95% at 4 hours, between about 45% and about 100% at 8 hours, greater than about 55% at 12 hours, greater than about 65% at 18 hours, and greater than about 70% at 24 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 °C of between 0% and about 40% at 1 hour, between about 0% and about 70% at 2 hours, between about 5% and about 95% at 4 hours, between about 12.5% and about 100% at 8 hours, between about 20% and about 100% at 12 hours, between about 35% and about 100% at 16 hours, between about 55% and about 100% at 24 hours, and greater than about 75% at 36 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% and about 60% at 1 hour, between about 0% and about 75% at 2 hours, between about 5% and about 95% at 4 hours, between about 12.5% and about 100% at 8 hours, between about 15% and about 100% at 12 hours, between about 25% to about 100% at 16 hours, between about 30% and about 100% hours at 24 hours and greater than 60% at 36 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) at 37 0C of between 0% to about 50% by weight of the abusable drug.
  • SGF Simulated Gastric Fluid
  • said release rate is between 0% to about 1%, or 0% to about 3%, or 0% to about 5%, or 0% to about 10%, or 0% to about 15%, or 0% to about 20%, 0% to about 30%, or 0% to about 40%, or 0% to about 60%, or 0% to about 70%, or 0% to about 80%, or 0% to about 90%, 0% to about 100%.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% and about 60% at 1 hour, between about 0% and about 80% at 2 hours, between about 3% and about 95% at 4 hours and between about 10% and about 100% at 8 hours.
  • said release rate is between 0% and about 10% at 1 hour, between about 0% and about 20% at 2 hours, between about 2% and about 80% at 4 hours and between about 5% and about 100% at 8 hours; or between 0% and about 20% at 1 hour, between about 0% and about 40% at 2 hours, between about 0% and about 80% at 4 hours and between about 2% and about 100% at 8 hours; or between 0% and about 40% at 1 hour, between about 0% and about 60% at 2 hours, between about 5% and about 85% at 4 hours and between about 5% and about 90% at 8 hours and greater than 20% at 12 hours; or between 0% and about 50% at 1 hour, between about 0% and about 50% at 2 hours, between about 10% and about 90% at 4 hours and between about 15% and about 90% at 8 hours and greater than 30% at 12 hours; -or between 0% and about 70% at 1 hour, between about 0% and about 70% at 2 hours, between about 10% and about 75% at 4 hours and between about 15% and about 90% at 8 hours and greater than 30% at 12 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 10% and about 65% at 1 hour, between about 20% and about 75% at 2 hours, between about 30% and about 95% at 4 hours and between about 40% and about 100% at 8 hours.
  • said release rate is between 2% and about 70% at 1 hour, between about 5% and about 80% at 2 hours, between about 10% and about 90% at 4 hours and between about 20% and about 100% at 8 hours; or between 5% and about 60% at 1 hour, between about 10% and about 75% at 2 hours, between about 15% and about 85% at 4 hours and between about 30% and about 100% at 8 hours; or between 20% and about 70% at 1 hour, between about 20% and about 75% at 2 hours, between about 20% and about 90% at 4 hours and between about 40% and about 100% at 8 hours; or between 30% and about 80% at 1 hour, between about 40% and about 85% at 2 hours, between about 40% and about 90% at 4 hours and between about 60% and about 100% at 8 hours; or between 1% and about 20% at 1 hour, between about 5% and about 20% at 2 hours, between about 10% and about 40% at 4 hours and between about 20% and about 40% at 8 hours and greater than 40% at 12 hours.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% ' to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours.
  • said release rate is between 0% to about 30% at 1 hour, from about 5% to about 45% at 2 hours, from about 10% to about 60% at 4 hours, from about 15% to about 70% at 6 hours, from about 25% to about 80% at 9 hours, and greater than about 50% at 12 hours; or between 0% to about 20% at 1 hour, from about 2% to about 35% at 2 hours, from about 5% to about 50% at 4 hours, from about 10% to about 60% at 6 hours, from about 15% to about 70% at 9 hours, and greater than about 40% at 12 hours; or between 0% to about 10% at 1 hour, from about 1% to about 30% at 2 hours, from about 5% to about 40% at 4 hours, from about 10% to about 60% at 6 hours, from about 15% to about 70% at 9 hours, and greater than about 40% at 12 hours; or between 0% to about 5% at 1 hour, from about 0% to about 10% at 2 hours, from about 2% to about 20% at 4 hours, from about 5% to about 30% at 6 hours, from about 10% to about 40% at 9 hours, and greater than about 50% at 12 hours
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between L6 and 7.2 at 37 0 C of between 5% and about 50% at 1 hour, between about 10% and about 75% at 2 hours, between about 20% and about 95% at 4 hours, between about 40% and about 100% at 8 hours, greater than about 50% at 12 hours, greater than about 70% at 18 hours, and greater than about 80% at 24 hours.
  • said release rate is between 2% and about 50% at 1 hour, between about 5% and about 75% at 2 hours, between about 15% and about 75% at 4 hours, between about 30% and about 90% at 8 hours, greater than about 40% at 12 hours, greater than about 60% at 18 hours, and greater than about 70% at 24 hours; or between 1% and about 40% at 1 hour, between about 2% and about 60% at 2 hours, between about 10% and about 65% at 4 hours, between about 20% and about 80% at 8 hours, greater than about 30% at 12 hours, greater than about 40% at 18 hours, and greater than about 60% at 24 hours; or between 5% and about 60% at 1 hour, between about 15% and about 80% at 2 hours, between about 25% and about 95% at 4 hours, between about 45% and about 100% at 8 hours, greater than about 60% at 12 hours, greater than about 80% at 18 hours, and greater than about 90% at 24 hours; or between 10% and about 65% at 1 hour, between about 20% and about 85% at 2 hours, between about 30% and about 100% at 4 hours, between about 60% and about 100% at 4 hours, between
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours.
  • said release rate is between 0% to about 20% at 1 hour, from about 5% to about 50% at 4 hours, from about 10% to about 60% at 8 hours, from about 15% to about 70% at 12 hours, from about 25% to about 90% at 18 hours, and greater than about 55% at 24 hours; or between 0% to about 10% at 1 hour, from about 5% to about 40% at 4 hours, from about 8% to about 50% at 8 hours, from about 10% to about 60% at 12 hours, from about 22% to about 80% at 18 hours, and greater than about 45% at 24 hours; or between 0% to about 35% at 1 hour, from about 15% to about 70% at 4 hours, from about 25% to about 75% at 8 hours, .
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 3% and about 95% at 4 hours, between about 10% and about 100% at 8 hours, between about 25% and about 100% at 12 hours, between about 30% and about 100% at 16 hours, between about 50% and about 100% at 24 hours, and greater than about 80% at 36 hours.
  • said release rate is between 0% and about 40% at 1 hour, between about 0% and about 65% at 2 hours, between about 2% and about 85% at 4 hours, between about 8% and about 90% at 8 hours, between about 20% and about 95% at 12 hours, between about 25% and about 95% at 16 hours, between about 40% and about 90% at 24 hours, and greater than about 70% at 36 hours; or between 0% and about 30% at 1 hour, between about 0% and about 50% at 2 hours, between about 1% and about 75% at 4 hours, between about 5% and about 80% at 8 hours, between about 10% and about 85% at 12 hours, between about 15% and about 90% at 16 hours, between about 30% and about 80% at 24 hours, and greater than about 70% at 36 hours; or between 0% and about 60% at 1 hour, between about 0% and about 80% at 2 hours, between about 5% and about 100% at 4 hours, between about 15% and about 100% at 8 hours, between about 35% and about 100% at 12 hours, between about 40% and about 100% at 16 hours, between about 60% and about 100% at
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 20% and about 50% at 1 hour, between about 40% and about 75% at 2 hours, between about 60% and about 95% at 4 hours, between about 80% and about 100% at 8 hours and between about 90% and about 100% at 12 hours.
  • said release rate is between 15% and about 45% at I hour, between about 35% and about 70% at 2 hours, between about 55% and about 90% at 4 hours, between about 75% and about 90% at 8 hours and between about 80% and about 95% at 12 hours; or between 10% and about 40% at 1 hour, between about 30% and about 65% at 2 hours, between about 50% and about 85% at 4 hours, between about 70% and about 85% at 8 hours and between about 75% and about 90% at 12 hours; or between 5% and about 35% at 1 hour, between about 25% and about 60% at 2 hours, between about 45% and about 80% at 4 hours, between about 65% and about 80% at 8 hours and between about 70% and about 85% at 12 hours; or between 25% and about 55% at 1 hour, between about 45% and about 80% at 2 hours, between about 65% and about 95% at 4 hours, between about 85% and about 100% at 8 hours and between about 95% and about 100% at 12 hours; or between 30% and about 60% at 1 hour, between about 50% and about 80% at 2 hours, between 30% and about 60% at
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 10% and about 95% at 4 hours, between about 35% and about 100% at 8 hours, between about 55% and about 100% at 12 hours, between about 70% to about 100% at 16 hours, and greater than about 90% at 24 hours.
  • said release rate is between 0% and about 40% at 1 hour, between about 0% and about 65% at 2 hours, between about 8% and about 85% at 4 hours, between about 30% and about 90% at 8 hours, between about 45% and about 100% at 12 hours, between about 60% to about 100% at 16 hours, and greater than about 80% at 24 hours; or between 0% and about 30% at 1 hour, between about 0% and about 55% at 2 hours, between about 5% and about 75% at 4 hours, between about 20% and about 80% at 8 hours, between about 35% and about 100% at 12 hours, between about 50% to about 100% at 16 hours, and greater than about 70% at 24 hours; or between 0% and about 20% at 1 hour, between about 0% and about 45% at 2 hours, between about 5% and about 65% at 4 hours, between about 10% and about 70% at 8 hours, between about 25% and about 80% at 12 hours, between about 40% to about 100% at 16 hours, and greater than about 60% at 24 hours; or between 0% and about 60% at 1 hour, between about 0% and about 80%
  • said release rate is between 0% and about 25% at 1 hour, between about 0% and about 40% at 2 hours, between about 2% and about 50% at 4 hours, between about 8% and about 60% at 8 hours, between about 10% and about 70% at 12 hours, between about 25% to about 80% at 16 hours, between about 45% and about 100% hours at 24 hours and greater than 75% at 36 hours; or between 0% and about 20% at 1 hour, between about 0% and about 35% at 2 hours, between about 1% and about 45% at 4 hours, between about 5% and about 55% at 8 hours, between about 8% and about 65% at 12 hours, between about 20% to about 75% at 16 hours, between about 40% and about 100% hours at 24 hours and greater than 70% at 36 hours; or between 0% and about 15% at 1 hour, between about 0% and about 30% at 2 hours, between about 0% and about 40% at 4 hours, between about 5% and about 50% at 8 hours, between about 8% and about 60% at 12 hours, between about 15% to about 70% at 16 hours, between about 35% and about 100% hours
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 3% and about 95% at 4 hours, between about 10% and about 100% at 8 hours, between about 20% and about 100% at 12 hours, between about 30% to about 100% at 16 hours, between about 50% and about 100% hours at 24 hours and greater than 80% at 36 hours, hi other preferred embodiments, said release rate is between 0% and about 45% at 1 hour, between about 0% and about 70% at 2 hours, between about 3% and about 90% at 4 hours, between about 8% and about 100% at 8 hours, between
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and ' 7.2 at 37 °C of between 15% and about 25% at 1 hour, between about 25% and about 35% at 2 hours, between about 30% and about 45% at 4 hours, between about 40% and about 60% at 8 hours, between about 55% and about 70% at 12 hours and between about 60% to about 75% at 16 hours.
  • said release rate is between 10% and about 20% at 1 hour, between about 20% and about 30% at 2 hours, between about 25% and about 40% at 4 hours, between about 30% and about 50% at 8 hours, between about 50% and about 65% at 12 hours and between about 55% to about 65% at 16 hours; or between 5% and about 15% at 1 hour, between about 15% and about 25% at 2 hours, between about 20% and about 35% at 4 hours, between about 25% and about 45% at 8 hours, between about 45% and about 60% at 12 hours and between about 50% to about 60% at 16 hours; or between 15% and about 30% at 1 hour, between about 20% and about 40% at 2 hours, between about 20% and about 50% at 4 hours, between about 30% and about 70% at 8 hours, between about 60% and about 80% at 12 hours and between about 70% to about 90% at 16 hours; or between 0% and about 50% at 1 hour, between about 5% and about 50% at 2 hours, between about 5% and about 70% at 4 hours, between about 10% and about 80% at 8 hours, between about 20% and about 100% at 12 hours and between about 40% to
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said in-vitro release rate being substantially independent of pH in that a difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH, when measured in-vitro using the USP Basket and Paddle Methods of USP Drug Release test of U.S. Pharmacopeia (2003) at 100 rpm in 900 ml aqueous buffer, is no greater than 30%.
  • the difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH using the aforementioned methods is no greater than 50%, or no greater than 40%, or no greater than 35%, or no greater than 25%, or no greater than 20%, or no greater than 15%, or no greater than 10%, or no greater than 5%.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said dosage forms of abusable drug providing in-vitro release rates by weight of between 0% to about 50% by weight of the abusable drug from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) at 37 °C.
  • SGF Simulated Gastric Fluid
  • said release rate at one hour is between 0% to about 10% by weight, or 0% to about 20% by weight, or is between 0% to about 30% by weight, or 0% to about 40% by weight, or between 0% to about 60% by weight, or 0% to about 70% by weight, or 0% to about 80% by weight, or 0% to about 90% by weight, or 10% to about 50% by weight, or 10% to about 60% by weight, or 10% to about 70% by weight, or 10% to about 90% by weight, or 10% to about 100% by weight, or 30% to about 100% by weight, or 50% to about 100% by weight.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said dosage forms of abusable drug providing in- vitro release rates by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0 C of between 0% to about 80% at 0.5 hours, and greater than about 40% at 1 hour.
  • said release rate is between 0% to about 40% at 0.5 hours, and greater than about 60% at 1 hour; or between 0% to about 20% at 0.5 hours, and greater than about 40% at 1 hour; or between 0% to about 20% at 0.5 hours, and greater than about 20% at 1 hour; or between 0% to about 90% at 0.5 hours, and greater than about 60% at 1 hour; or between 0% to about 100% at 0.5 hours, and greater than about 60% at 1 hour; or between 0% to about 90% at 1 hour, and greater than about 40% at 2 hours; or between 0% to about 100% at 1 hour, and greater than about 60% at 2 hours; or between 0% to about 60% at 1 hour, and greater than about 40% at 2 hours; or between 0% to about 40% at 1 hour, and greater than about 30% at 2 hours; or between 0% to about 50% at 1 hour, and greater than about 40% at J 2 hours; or between 0% to about 30% at 1 hour, and greater than about 20% at 2 hours; or between 0% and about 50% at 1 hour, between about 0% and about 80% at
  • the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug . and (ii) ADER, such that the ratio of the mean C max of the abusable drug after single dose oral administration of the dosage form after tampering to the mean C max of abusable drug after single dose oral administration of an intact dosage form is not more than about 20: 1.
  • the mean C max ratio using the aforementioned test method is not more than about 15:1, or about 10:1, or about 7.5:1, or about 6:1, or about 5:1, or about 4:1, or about 3:1, or about 2:1, or about 1.5:1, or about 1.25:1.
  • the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean C max of the abusable drug after single dose oral administration of an immediate release reference product containing an equivalent amount of abusable drug to the mean C ma ⁇ of abusable drug after single dose oral administration of an intact dosage form of the invention is at least about 1.25:1.
  • the mean C max ratio using the aforementioned test method is at least about 1.5:1, or about 2:1, or about 3:1, or about 4:1, or about 5:1, or about 6:1, or about 10:1, or about 15:1 or about 20:1.
  • the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean AUC0-2 of the abusable drug after single dose oral administration of the dosage form after tampering to the mean AUC 0 -2 of abusable drug after single dose oral administration of an intact dosage form is not more than about 20:1.
  • the mean AUC ratio using the aforementioned test method is measured from time 0 to up to 1, 2.5, 3, 4, 5 or 6 hours post dose (i.e., AUCo -1 , AUCo-2.5, AUC0-3, AUCo-4, AUC 0- S and AUC 0 - 6 , respectively).
  • the mean AUCo-i, AUC0-2, AUCo-2.5, AUC0-3, AUC0-4, AUC0-5 and AUCo- 6 ratios using the aforementioned test method are not more than about 15:1, or about 10:1, or about 7.5:1, or about 6:1, or about 5:1, or about 4:1, or about 3:1, or about 2:1 or about 1.5:1.
  • the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean AUCo-2 of the abusable drug after single dose oral administration of an immediate release reference product containing an equivalent amount of abusable drug to the mean AUC 0-2 of abusable drug after single dose oral administration of an intact dosage form of the invention is at least about 1.25:1.
  • the mean AUC ratio using the aforementioned test method is measured from time 0 to up to 1, 2.5, 3, 4, 5 or 6 hours post dose (i.e., AUC 0- I, AUCo-2.5, AUC 0-3 , AUC0-4, AUCo-5 and AUCo-6, respectively).
  • the mean AUC 0-1 , AUC 0-2 , AUCo-2.5, AUC0-3, AUC 0-4 , AUC 0 . 5 and AUC 0-6 ratios using the aforementioned test method are not more than about 15:1, or about 10:1, or about 7.5:1, or about 6:1, or about or about 5:1, or about 4:1, or about 3:1, or about 2:1 or about 1.5:1.
  • the oral dosage forni of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean T ma ⁇ of the abusable drug after single dose oral administration of the intact dosage form to the mean T max of abusable drug after single dose oral administration of an dosage form after tampering is not more than about 20:1.
  • the mean T ma ⁇ ratio using the aforementioned test method is not more than about 15:1, or not more than about 10:1, or not more than about 7.5:1, or not more than about 6:1, or not more than about 5:1, or not more than about 4:1, or not more than about 3:1, or not more than about 2:1, or not more than about 1.5:1, or not more than about 1.25:1.
  • the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean T max of the abusable drug after single dose oral administration of an immediate release reference product containing an equivalent amount of abusable drug to the mean Tmax of abusable drug after single dose oral administration of an intact dosage form of the invention is at least about 1.25:1.
  • the mean T max ratio using the aforementioned test method is at least about 1.5:1, or at least about 2: 1, or at least about 3:1, or at least about 4:1, or at least about 5:1, or at least about 6:1, or at least about 10:1, or at least about 15:1 or at least about 20:1.
  • the invention is directed to an oral dosage form comprising (i) an abusable drug and (ii) ADER, such that less than 70% of the abusable drug is released from the intact dosage form after 1 hour based on the in- vitro dissolution of the dosage form in 900 mL of 40% ethanol in water using the USP Basket and Paddle Methods at 50 rpm and 37°C.
  • the release rate of the abusable drug from the intact dosage form by the aforementioned USP basket method at 1 hours is 60% or less, 50% or less, 45% or less, 40% or less, 35% or less, 33% or less, 30% or less, 25% or less, 20% or less or 15% or less.
  • the mean ratio of the amount of abusable drug released from the dosage form after mechanical tampering e.g., after crushing with a single crush of a spatula or in the case of a capsule containing a solid, cutting into two pieces
  • the amount of abusable drug released from the intact dosage form based on the dissolution at 0.5 hours of the dosage form in 900 mL of Simulated Gastric Fluid using the USP Basket and Paddle Methods at 50 rpm at 37 degrees °C is less than 20:1.
  • the mean ratio by the aforementioned USP basket method at 0.5 hours is 15:1 or less, 10:1 or less, 7.5:1 or less, 5:1 or less. 3:1 or less, 2:1 or less, 1.5:1 or less.
  • the mean ratio of the amount of abusable drug released from the dosage form after mechanical tampering e.g., after crushing with a single crush of a spatula or in the case of a capsule containing a solid, cutting into two pieces
  • the mean ratio by the aforementioned USP basket method at 1 hour is 15:1 or less, 10:1 or less, 7.5:1 or less, 5:1 or less. 3:1 or less, 2:1 or less, 1.5:1 or less.
  • the mean ratio of the amount of abusable drug released from the dosage form after mechanical tampering e.g., after crushing with a single crush of a spatula or in the case of a capsule containing a solid, cutting into two pieces
  • the mean ratio by the aforementioned USP basket method at 2 hours is 15:1 or less, 10:1 or less, 7.5:1 or less, 5:1 or less. 3:1 or less, 2:1 or less, 1.5:1 or less.
  • the present invention is directed to an oral dosage form comprising (i) an abusable drug and (ii) ADER, such that the ratio of the mean C m ax of the abusable drug after single dose oral administration of the dosage form after tampering to the mean C max of abusable drug after single dose oral administration of an intact dosage form is less than about 20:1.
  • said mean ratio using the aforementioned test method is less than about 15:1 or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 4:1, or less than about 3:1, or less than 2.5:1, or less than about 2:1, or less than about 1.75:1, or less than about 1.5:1, or less than about 1.25:1 or less than about 1.25:1
  • the present invention is directed to an oral dosage form comprising (i) an abusable drug and (ii) ADER, such that the ratio of the mean AUCo -2 of the abusable drug after single dose oral administration of an immediate release dosage form containing an equivalent amount of abusable drug to the mean AUCo- 2 of abusable drug after single dose oral administration of an intact dosage form of the invention is at least 1.25:1.
  • the mean AUC0-2 ratio using the aforementioned test method is at least about 1.5:1, or at least about 1.75:1, or at least about 2:1, or at least about 2.5:1, or at least about 3:1, or at least about 3.5:1, or at least about 4:1, or at least about 5:1, or at least about 6:1, or at least about 10:1 or at least about 15;1 or at least about 20:1.
  • the invention is also directed to methods of preventing abuse and misuse of an abusable drug utilizing the dosage forms disclosed herein.
  • the method can comprise providing the abusable drug in an oral dosage form together with ADER, wherein the abusable drug is present in a form which is partially or substantially resistant to tampering (e.g., crushing, shear forces which break up the dosage form, solvent extraction, etc.).
  • tampering e.g., crushing, shear forces which break up the dosage form, solvent extraction, etc.
  • the release for the abusable drug component of the formulation is expressed in terms of a ratio of the release achieved after tampering, relative to the amount released from the intact formulation.
  • the ratio is therefore expressed as [Crushed]/[Whole], and it is desired that this ratio have a numerical range of not more than 20:1 (crushed release in 1 hour/intact release in 1 hour), based on in-vitro dissolution of the dosage form in 900 ml of Simulated Gastric Fluid using the USP Basket and Paddle Methods at 50 rpm and 37°C.
  • the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5 : 1 , or less than about 1.25 : 1.
  • the abusable drug dosage form of the invention is bioequivalent when taken under fed and fasted conditions.
  • the abusable drug dosage form of the invention upon administration provides a mean fed to fasted abusable drug AUCo- ⁇ difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%.
  • the abusable drug dosage form of the invention upon administration provides a mean fed to fasted abusable drug Cm 0x difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%.
  • the abusable drug dosage form of the invention is bioequivalent when taken under with and without 30 mL, 60 mL, 90 mL, 120 mL, 180 mL, 240 mL, 270 mL, 300 mL of a 40% ethanol solution.
  • the abusable drug dosage form of the invention upon administration provides a mean alcohol to no alcohol state abusable drug AUCo- ⁇ difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%, said alcohol state induced by concurrent ingestion of the abusable drug with about 300 mL or about 270 mL, or about 240 mL, or about 210 mL, or about 180 mL or about 150 mL, or about 120 mL, or about 90 mL, or about 60 mL or about 30 mL of a 40% solution of ethanol.
  • the abusable drug dosage form of the invention upon administration provides a mean alcohol to no alcohol state abusable drug C max difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%, said alcohol state induced by concurrent ingestion of the abusable drug with about 300 mL or about 270 mL, or about 240 mL, or about 210 mL, or about 180 mL or about 150 mL, or about 120 mL, or about 90 mL, or about 60 mL or about 30 mL of a 40% solution of ethanol.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof, ADER material, and a pharmacologic antagonist to the abusable drug.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof, ADER material, and a pharmacologic antagonist to a co-abused abusable drug, said co-abused abusable drug not part of the dosage form.
  • the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof, ADER material, and a pharmacologic antagonist for both the abusable drug of the dosage form and a co-abused abusable drug, said co-abused abusable drug not part of the dosage form.
  • the dosage form of the invention comprises a therapeutically effective amount of abusable drug or pharmaceutically acceptable salt thereof or mixture thereof, ADER material, and a pharmacologic antagonist to the abusable drug (and/or to a co-abused abusable drug, said co-abused abusable drug not part of the dosage form)
  • some or all of the in vitro dissolution rates or in-vitro release rates e.g., USP Basket and Paddle Methods
  • USP Basket and Paddle Methods e.g., USP Basket and Paddle Methods
  • the in vitro dissolution rates or in-vitro release rates for the abusable drug and the pharmacologic antagonists are the same. In other preferred embodiments, the in vitro dissolution rates or in-vitro release rates for the abusable drug and for the pharmacologic antagonists are from different.
  • the dosage form of the invention comprises a therapeutically effective amount of abusable drug or pharmaceutically acceptable salt thereof or mixture thereof, ADER material, and an aversive agent
  • some or all of the in vitro dissolution rates or in-vitro release rates e.g., USP Basket and Paddle Methods
  • the in vitro dissolution rates or in-vitro release rates for the abusable drug and the aversive agent are the same.
  • the in vitro dissolution rates or in-vitro release rates for the abusable drug and for the aversive agent are different.
  • the dosage form of the invention comprises a therapeutically effective amount of abusable drug or pharmaceutically acceptable salt thereof or mixture thereof, ADER material, and a pharmacologic antagonist to the abusable drug (and/or to a co-abused abusable drug, said co-abused abusable drug not part of the dosage form)
  • some or all of the pharmacokinetic parameters e.g., AUCo- t , AUCo--, AUCo- 8, AUC 0 -I 2 , AUC 0-24 , C max , C 8 , C 12 , C 24 , W or T max , C min , HVD, W 50 , steady state, percent fluctuation and AI
  • AUCo- t AUCo- t
  • AUCo-- AUCo- 8
  • AUC 0 -I 2 e.g., AUC 0-24 , C max , C 8 , C 12 , C 24 , W or T max , C min , HVD
  • the pharmacokinetic parameters for the abusable drug and the pharmacologic antagonists are the same. In other preferred embodiments, the pharmacokinetic parameters for the abusable drug and for the pharmacologic antagonists are different.
  • the ADER material provides extended release of the abusable drug and the pharmacologic antagonist for the abusable drug, thereby significantly reducing the risk precipitating signs and symptoms of abusable drug withdrawal or an abstinence syndrome in abusable drug dependent and tolerant individuals, and diminution of the intended therapeutic response from the abusable drug.
  • the ADER material provides one line of defense and the antagonist provides yet another line of defense against drug abuse.
  • the amount of pharmacologic antagonist to the abusable drug in the oral dosage form will vary for a variety of reasons, including the choice of abusable drug, the potency of the abusable drug, the potency of the antagonist, the oral bioavailability of the abusable drug and the antagonist, the safety and tolerability of the antagonist, the degree of blockade sought to the effects of the abusable drug, the patients prior exposure to the abusable drug, the nature of the formulation (e.g., immediate release or extended release), the pharmacokinetics, pharmacodynamics and physicochemical characteristics of the abusable drug and the antagonist.
  • pharmacologic antagonist to the abusable drug is about 0.00001 mg to 1000 mg, or about 0.00001 mg to about 700 mg, or about 0.001 mg to about 500 mg, or about 0.01 mg to about 400 mg, or about 0.1 mg to about 200 rag, or about 1 mg to about 100 mg, or about 0.00001 to about 500 mg, or about 0.001 to about 300 mg, or about 0.01 to about 200 mg.
  • the mean ratio of the time to confirmed perceptible pain relief after administration of the intact dosage form to the time to confirmed perceptible pain relief after administration of the tampered dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 15:1, .or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the mean ratio of the time to meaningful pain relief after administration of the intact dosage form to the time to meaningful pain relief after administration of the tampered dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2: 1 , or less than about 1.5 : 1 , or less than about 1.25 : 1.
  • the mean ratio of the peak pain intensity difference score after administration of the tampered dosage form to the peak pain intensity difference score after administration of the intact dosage form is less than 10:1.
  • the mean ratio using the aforementioned test method is less than about 8:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the mean ratio of the peak pain relief score after administration of the tampered dosage form to the peak pain relief score after administration of the intact dosage form is less than 10:1.
  • the mean ratio using the aforementioned test method is less than about 8:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the mean ratio of change from baseline to two hours post-dose in pain intensity score after administration of the tampered dosage form to the change from baseline to two hours post-dose in pain intensity score after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the mean ratio of the number of patients with pain who need to be treated to obtain > 50% pain relief in one patient (i.e., number needed to treat or NNT) after administration of the tampered dosage form to the NNT after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the aforementioned at NNT is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post- dose.
  • the mean ratio of the number needed to harm (referred to hereinafter as "NNH") due to moderate or severe nausea in healthy subjects (naive to said analgesic) after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post- dose.
  • the mean ratio of the NNH due to moderate or severe sedation or drowsiness in healthy subjects (na ⁇ ve to said analgesic) after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is lessMhan about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to moderate or severe sedation or drowsiness in healthy subjects who are na ⁇ ve to said abusable drug and who are occasional or light consumers of alcohol after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1, said NNH measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
  • the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
  • the mean ratio of the number of patients who need to be treated (NNT) to obtain > 50% reduction in the cardinal sign or symptom of the medical condition (e.g., pain relief when the medical condition is pain, attention deficit hyperactivity disorder-rating scale [ADHD-RS-IV total scores] when the medical condition is ADHD, etc.) after administration of the tampered dosage form to the NNT after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNT is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to moderate or severe sedation or drowsiness in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to moderate or severe sedation or drowsiness in healthy subjects who are na ⁇ ve to said abusable drug and who are occasional or light consumers of alcohol, after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1, said NNH measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1 , or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the mean ratio of the NNH due to moderate or severe nausea in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the abusable drug produces CNS stimulation
  • the abusable drug is a stimulant, CNS-stimulant, psychostimulant, alkylxanthine or anorectic
  • the mean ratio of the NNH due to the incidence of tachycardia i.e., resting heart rate > 100 beats per minute
  • the mean ratio of the NNH due to the incidence of tachycardia i.e., resting heart rate > 100 beats per minute
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1 , or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the abusable drug produces CNS stimulation (e.g., the abusable drug is a stimulant, CNS-stimulant, psychostimulant, alkylxanthine or anorectic) upon administration to na ⁇ ve (i.e., abusable drug na ⁇ ve) healthy subjects
  • the mean ratio of the NNH due to the incidence of palpitations in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16: 1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to the incidence of headache in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1 , or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to the incidence of dizziness in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15: 1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to the incidence of lightheadedness in naive healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1 :1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to the incidence of dry mouth in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1 :1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the NNH due to the incidence of insomnia in na ⁇ ye healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the mean ratio of the NNH due to the incidence of abdominal pain or abdominal discomfort in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1 , 1.5, 2, 3, 4, 5 or 6 hours post-dose
  • the mean ratio of the NNH due to the incidence of nervousness in na ⁇ ve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the drug liking score after administration of the tampered dosage form to the mean ratio of the drug liking score after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose
  • the mean ratio of the drug effect score after administration of the tampered dosage form to the mean ratio of the drug effect score after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1 , or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the score on the "take again” questionnaire after administration of the tampered dosage form to the mean ratio of the score on the "take again” questionnaire after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of the score on the "coasting" questionnaire after administration of the tampered dosage form to the mean ratio of the score on the "coasting” questionnaire after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of impairment on the "critical tracking task" driving skills test in na ⁇ ve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of impairment on the "critical tracking task" driving skills test score in na ⁇ ve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1, said "critical tracking task” driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1 , or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the mean ratio of impairment on the "stop signal task" driving skills test in na ⁇ ve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1.
  • the mean ratio of impairment on the "stop signal task" driving skills test in na ⁇ ve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of impairment on the "stop signal task” driving skills test score in na ⁇ ve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1, said "stop signal task” driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1 :1.
  • the mean ratio of impairment on the "Tower of London" driving skills test score in na ⁇ ve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
  • the mean ratio of impairment on the "Tower of London” driving skills test score in na ⁇ ve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1 said "Tower of London” driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about L5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
  • the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
  • the invention is also directed to a method of treating or preventing diseases and disorders amenable to treatment with abusable drugs.
  • the method can comprise providing an oral dosage form containing an abusable drug and ADER, said dosage form an immediate release formulation, an extended release formulation or a formulation comprising both immediate release and extended release.
  • ADER includes, but is not limited to tablets or capsules.
  • the dosage forms of the present invention may include any desired pharmaceutical excipients known to those skilled in the art.
  • the oral dosage forms may further provide an immediate release of the abusable drug, hi certain preferred embodiments, the oral dosage forms of the present invention provide a sustained release of the abusable drug contained therein.
  • Oral dosage forms providing sustained release of the abusable drug may be prepared in accordance with formulations/methods of manufacture known to those skilled in the art of pharmaceutical formulation.
  • the benefits of the abuse-resistant dosage form are especially great in connection with oral dosage forms of potent abusable drug, which can provide valuable therapeutic benefits but are prone to being abused. This is particularly true for sustained release abusable drug products which have a large dose of a desirable abusable drug intended to be released over a period of time in each dosage unit. Drug abusers take such sustained-release product and crush, grind, extract or otherwise damage the product so that the full contents of the dosage form become available for immediate absorption. Since such tampering of the dosage form of the invention results in the abusable drug also becoming available for absorption, the present invention provides a means for deterring such abuse.
  • the present invention addresses the risk of overdose to non-abusing patients from "dumping" effect of the full dose of the abusable drug if the product is accidentally chewed or crushed, co-ingested with a significant amount of alcohol or taken interchangeably in fasted and fed states.
  • a combination of two abusable drug is included in the formulation with the ADER.
  • one or more abusable drug and ADER are included and a further non-abusable drug is also included for the treatment of the same medical condition as the abusable drug or for the treatment of a different medical condition.
  • Another embodiment of the invention is directed to a method of preventing or treating pain, addiction disorders, spasticity, musculoskeletal disorders, ADHD, insomnia, excessive sleepiness, daytime sleepiness, sleep disorders, anxiety disorders, panic attacks, agoraphobia, obsessive-compulsive disorders, psychiatric disorders, neurologic disorders, excess weight, obesity and other medical maladies responsive to treatment with the abusable drugs with the disclosed dosage forms.
  • the method of treating the aforementioned disorders in patients with a dosage form having less abuse potential comprises providing an oral dosage form containing an abusable drug and ADER; and orally administering the dosage form to provide a plasma level of abusable drug greater than the minimum therapeutic or minimum effective concentration of the abusable drug.
  • the invention is also directed to methods of preparing the dosage forms disclosed herein.
  • the benefits of the abuse-resistant dosage form are especially great in connection potent abusable drugs, which would provide valuable therapeutic benefits but would be prone to being abused. This is particularly true for oral dosage forms, including, in some preferred embodiments, sustained release dosage forms of abusable drugs which would have a large dose of a desirable drug intended to be released over a period of time in each dosage unit. Drug abusers may tamper the dosage form of the invention so that the full contents of the dosage form become available for immediate and maximal mood altering effects.
  • the dosage form of the present invention would reduce the mood altering effects of the abusable drugs upon tampering and as such the invention provides pharmaceutical compositions, dosage forms and methods of deterring misuse, abuse, tampering and diversion of the dosage form.
  • the dosage form of the invention when the dosage form of the invention is tampered, the amount of abusable drug released in immediate release form is reduced, which in turn reduces the euphoric, pleasurable, reinforcing, rewarding, mood altering and toxic effects of the abusable drugs of the dosage form.
  • the dosage form of the present invention When the dosage form of the present invention is orally administered as intended to humans, the abusable drugs is released into systemic circulation as intended and is therefore available for absorption into the body. However, if ' the dosage forms of the present invention is tampered (e.g., chemical, solvent, thermal or mechanical extraction, followed by administration into the body) the ADER of the invention would reduce the amount of abusable drugs available in immediate release form.
  • the dosage form of the invention substantially reduces the efficiency of drug aspiration into syringes, drug filtration after solvent extraction and drug extraction after attempts at chemical, mechanical or thermal extraction from both immediate and sustained release dosage form of the invention. These characteristic decrease the potential for abuse or diversion of the abusable drugs in the dosage form by blocking the mood altering, euphoric, pleasurable, reinforcing, rewarding or toxic effects of the abusable drug.
  • tampering means any manipulation by mechanical, thermal and/or chemical means which changes the physical or chemical properties of the dosage form, e.g., to liberate the abusable drugs for immediate release if it is in sustained release form, or to make the abusable drugs available for inappropriate use such as administration by an alternate route, e.g., parenterally.
  • the tampering can be, e.g., by means of crushing, shearing, grinding, mechanical extraction, solvent extraction, solvent immersion, combustion, heating or any combination thereof.
  • abuse includes intermittent use, recreational use and chronic use of abusable drugs alone or in conjunction with other drugs means use: (i) in quantities or by methods and routes of administration that do not conform to standard medical practice; (ii) outside the scope of specific instructions for use provided by a qualified medical professional; (iii) outside the supervision of a qualified medical professional; (iv) outside the approved instructions on proper use provided by the drug's legal manufacturer; (v) which is not in specifically approved dosage forms for medical use as pharmaceutical agents; (vi) where there is an intense desire for and efforts to procure same; (vii) compulsive use; (viii) through acquisition by manipulation of the medical system, including falsification of medical history, symptom intensity, disease severity, patient identity, doctor shopping, prescription forgeries; (
  • microood altering is defined for purposes of the present invention to mean that the "high”, “liking”, pleasurable, euphoric, alerting, calming, anxiolytic, auditory and visual perceptual alterations, relaxing, psychotomimetic, rewarding, reinforcing and toxic effects of the abusable drug.
  • “deter abuse” and “deter abuse” are used interchangeably in the context of the present invention and include pharmaceutical compositions and methods that resist, deter, discourage, diminish, delay and/or frustrate: (i) the intentional, unintentional or accidental physical or chemical manipulation or tampering of the dosage form (e.g., crushing, shearing, grinding, chewing, dissolving, melting, needle aspiration, inhalation, insufflation, extraction by mechanical, thermal and chemical means, and/or filtration); (ii) the intentional, unintentional or accidental use or misuse of the dosage form outside the scope of specific instructions for use provided by a qualified medical professional, outside the supervision of a qualified medical professional and outside the approved instructions on proper use provided by the drug's legal manufacturer (e.g., intravenous use, intranasal use, inhalational use and oral ingestion to provide high peak concentrations); (iii) the intentional
  • aversive agents means to compounds contained within the dosage form that produce an aversive, undesirable, repugnant, distasteful, unpleasant, unacceptable physiologic or unacceptable psychic effects, or that pharmacologically block or reduce one or more of the following effects: mood alterations; euphoria, pleasure; a feeling of high; a feeling of drug liking; anxiolysis; mental stimulation; increased mental arousal; sedation; calmness; a state of relaxation; psychotomimesis; hallucinations; alterations in perception, cognition and mental focus; insomnia; hypersomnia; increased wakefulness or alertness; memory improvement; increased sexual gratification; increased sexual arousal; increased sexual desire and sexual anticipation; increased socialization; reduced social anxiety; psychologically reinforcement; and psychologically rewarding.
  • the intention of the aversive agent is deter or further deter the misuse, abuse, tampering or diversion of the drug for use by addicts, drug abusers and recreational drug users.
  • the aversive agent produces aversive effects only when the dosage form of the abusable drug of the invention is abused.
  • the aversive agent is contained within the dosage form at a dose or in a form that does not produce aversive effects when the dosage form of the abusable drug of the invention is taken as medically directed or in a manner that is consistent with the manufacturer's prescribing information, but which when abused or tampered with, produces an aversive effect.
  • the dosage form of the abusable drug of the invention contains one or more aversive agents in a non- releasable form (i.e., sequestered) form, said aversive agent partially or substantially released upon tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like).
  • aversive agents in a non- releasable form (i.e., sequestered) form, said aversive agent partially or substantially released upon tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like).
  • the dosage form of the abusable drug of the invention contains one or more aversive agents in releasable or partially releasable form, said dosage form not aversive when taken at medically approved doses or at doses consistent with the manufacturers prescribing information, said dosage form producing an aversive effect when taken in excess of medically approved doses or the manufacturers prescribing information.
  • the dosage form of the abusable drug of the invention contains one or more aversive agents in a non-releasable form (i.e., sequestered) form, said aversive agent partially or substantially released upon tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like), and said aversive agent pharmacologically blocking the effects of the abusable drug and/or the effects of a co-abused drug, said co-abused drug not part of the dosage form of the invention.
  • aversive agents in a non-releasable form (i.e., sequestered) form
  • said aversive agent partially or substantially released upon tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like)
  • said aversive agent pharmacologically blocking the effects of the abusable drug and/or the effects of
  • to qualify as an "ADER” requires a mixture of two or more compounds from the form the foregoing group [i.e., (a) to (d)]. In some particularly preferred embodiments, to qualify as an "ADER” requires a mixture of two or more compounds selected from at least two categories[i.e., (a) to (d)].
  • the dosage form may optionally also contain hydrophobic polymers, hydrophilic polymers, gums, protein derived materials, other waxes, shellac, other oils and mixtures thereof.
  • the invention is directed at an abusable drug dosage form, said dosage form having flotation capabilities to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage).
  • the invention is directed at an abusable drug dosage form, said dosage form having flotation capabilities to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage), said dosage form staying substantially afloat (i.e., floatable or buoyant) on a liquid surface for at least about 3 minutes.
  • afloat i.e., floatable or buoyant
  • said dosage form stays substantially afloat for at least about 5 minutes, or at least about 7 minutes, or at least about 10 minutes, or at least about 15 minutes, or at least about 20 minutes, or at least about 25 minutes, or at least about 30 minutes, or at least about 40 minutes, or at least about 50 minutes, or at least about 60 minutes, or at least about 90 minutes, or at least about 120 minutes, or at least about 150 minutes, or at least about 180 minutes, or at least about 210 minutes, or at least about 240 minutes, or at least about 270 minutes, or at least about 300 minutes, or at least about 330 minutes, or at least about 360 minutes, or at least about 400 minutes.
  • the invention is directed at an abusable drug dosage form, said dosage form having a non-toxic dye to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage).
  • the invention is directed at an abusable drug dosage form, said dosage form having a non-toxic bittering agent to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage).
  • the invention is directed at an abusable drug dosage form, said dosage form having a non-toxic bittering agent to deter oral or nasal ingestion of the dosage form.
  • the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined from first administration. In other preferred embodiments, the in vivo pharmacokinetic .parameters are derived or determined from steady state administration.
  • the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined under fed conditions. In other preferred embodiments, the in vivo pharmacokinetic parameters are derived or determined under fasted conditions.
  • the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined from an individual subject. In other preferred embodiments, the in vivo pharmacokinetic parameters are derived or determined from a population of subjects.
  • BMI Body Mass Index
  • BMI Body Mass Index
  • All pain states are contemplated by this invention, regardless of etiology, mechanisms, duration, prior treatment response and anatomic location, including acute pain, inflammatory pain, chronic pain, cancer pain, visceral pain and neuropathic pain.
  • kits for providing relief in a human patient suffering from neuropathic and chronic pain comprising a therapeutically effective amount of oral abusable drug which possesses analgesic properties or pharmaceutically acceptable salts thereof or mixtures thereof.
  • the dosage form of the invention is intended for the treatment of neuropathic pain, peripheral neuropathic pain, central neuropathic pain, chronic pain, osteoarthritis, back pain, cancer pain, fibromyalgia, and chronic inflammatory pain.
  • Also disclosed are methods of providing relief in a human patient suffering from acute pain comprising a therapeutically effective amount of oral abusable drug which possesses analgesic properties or pharmaceutically acceptable salts thereof or mixtures thereof.
  • kits of the present invention are contemplated.
  • kits for use in treating or preventing the pain with the oral administration of an abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof for a subject in need of such treatment comprising: (i) a dosage form of the invention; (ii) a container for the dosage form; and optionally, any of (iii) to (vi): (iii) a container for individual units of the dosage form (e.g., individual tablets or capsules in blisters); (iv) educational instructions in any media about various medical conditions, including without limitation, pain, ADHD, sleep disorders, anxiety disorders, obesity, neurologic disorders and psychiatric disorders, their etiology, pathophysiology, consequences and treatment, including information on the potential for abuse and diversion and methods for prevention of same and information on the proper use and disposal of the medication; (v) containers or bags for the safe disposal of any used or remaining unused dosage form, preferably child proof and flushable; (vi) tamper evident and child proof packaging for the
  • the amount of abusable drug in the oral dosage form will vary depending on variety of physiologic, pharmacologic, pharmacokinetic, pharmaceutical and physicochemical factors, including: (i) the choice of abusable drug as the unsalified form, pharmaceutically acceptable salt or mixtures therof; (ii) the nature of the oral dosage form (e.g, immediate release or extended release); (iii) the intensity and intractability of the medical condition; (iv) the absorption, metabolism, distribution and excretion of orally administered abusable drug in healthy subjects and in patients with various diseases and disorders, including renal and hepatic impairment; (v) the presence of comorbid pathology; (vi) the patient's risk of iatrogenic side effects; (vii) the tolerability of the dose, including the patient's propensity for abusable drug associated side effects; (viii) use of other drugs to treat the same medical condition; (ix) the efficiency of the dosage form; (x) the physicochemical properties of
  • the invention is also directed to methods of preparing the dosage forms disclosed herein.
  • the abusable drug in the dosage form is combined with one or more other drugs for the treatment of the same medical condition as the abusable drug or for the treatment of a different medical condition.
  • All modes of co-administration are contemplated, including via an oral, subcutaneous, direct intravenous, slow intravenous infusion, continuous intravenous infusion, intravenous or epidural patient controlled analgesia (PCA and PCEA), intramuscular, intrathecal, epidural, intracisternal, intramuscular, intraperitoneal, transdermal, topical, transmucosal, buccal, sublingual, transmucosal, inhalation, intranasal, epidural, intra-atricular, intranasal, rectal or ocular routes.
  • first administration means administration of a dose of the present invention at the initiation of therapy to an individual patient or a patient population.
  • steady state means that the amount of the drug reaching the system is approximately the same as the amount of the drug leaving the system.
  • the patient's body eliminates the drug at approximately the same rate that the drug becomes available to the patient's system through absorption into the blood stream.
  • AUCo- t means area under the plasma drug concentration-time curve from time zero to the "t", where t is the time point of the maximum intended dosing frequency of the dosage form (e.g., 4 hours, 6 hours, 8 hours, 12 hours or 24 hours for dosage forms intended to be administered every 4 hours, every 6 hours, every 8 hours, every 12 hours and every 24 hours, respectively, thereby providing an AUCo-t time interval of 0 to 4 hours, 0 to 6 hours, 0 to 8 hours, 0 to 12 hours and 0 to 24 hours, respectively);
  • "AUCo- ⁇ ” means area under the plasma drug concentration- time curve from time zero to infinity;
  • AUCo-s means area under the plasma drug concentration-time curve from time zero to 8 hours after dosing;
  • AUCo-i 2 means area under the plasma drug concentration-time curve from time zero to 12 hours after dosing;
  • AUCo -24 means area under
  • Pharmacokinetic parameters of the invention are be computed from first administration and steady state pharmacokinetic studies conducted in an individual subject or in a population of subjects in the fasted or fed states.
  • the AI and percent of steady state computations requires both single dose (i.e., first administration) and steady state pharmacokinetic assessment.
  • an effective amount of abusable drug in immediate release form is included in the controlled release unit dose abusable drug formulation to be administered.
  • the immediate release form of the abusable drug is preferably included in an amount which is effective to shorten the time to C max or increase the magnitude of the C max of the abusable drug in the blood (e.g., plasma).
  • an effective amount of the abusable drug in immediate release form may be coated onto the substrates of the present invention. For example, where the extended release abusable drug from the formulation is due to a controlled release coating, the immediate release layer would be overcoated on top of the controlled release coating.
  • the immediate release layer maybe coated onto the surface of substrates wherein the abusable drug is incorporated in a controlled release matrix.
  • the immediate release portion of the abusable drug dose may be incorporated into the gelatin capsule via inclusion of the sufficient amount of immediate release abusable drug as a powder or granulate within the capsule.
  • the gelatin capsule itself may be coated with an immediate release layer of the abusable drug.
  • the immediate release abusable drug is in liquid form, for example as a capsule within a capsule or as a liquid in contact with an extended release dosage form within a capsule.
  • any one or all of the above in- vivo parameters are achieved after a first administration (often referred to as "single dose administration") of the dosage form to a human patient or a population of human patients.
  • any one or all of the above in-vivo parameters are achieved after steady state administration of the dosage form to a human patient or a population of human patients.
  • Meaningful Pain Relief are assessed and defined as follows: At the time of dosing with the study medication, a trained member of study staff starts two stopwatches for each patient. The patient is instructed to stop the first stopwatch at the time of perceptible pain relief and the second stopwatch at the time when they first experience meaningful pain relief.
  • the usual definitions of the perceptible and meaningful pain relief are as follows: Perceptible Pain Relief is when the patient begins to feel any pain relieving effect from the drug. The patient is typically instructed as follows: "I would like you to stop the first stopwatch when you first feel any pain relief whatsoever. This does not mean you feel completely better, although you might, but when you first feel any difference in the pain that you have had”. Meaningful Pain Relief is when the patient feels their pain relief is meaningful to them.
  • the patient is typically instructed as follows: "I would like you to stop the second stopwatch when you have meaningful pain relief. That is, when the relief from the pain is meaningful to you". Confirmed Perceptible Pain Relief is Perceptible Pain Relief in those patients who go on to also have Meaningful Pain Relief.
  • NNT or “the number needed to treat” is the number of patients who need to be treated in order for one patient to obtain > 50% reduction in signs or symptoms (e.g., > 50% reduction in pain intensity or ADHD-RS-IV score).
  • the "NNH” or “number needed to harm” is a measure that indicates how many patients would require a specific treatment to cause harm in one patient.
  • the "NNH or “number needed to harm” is a measure that indicates: (i) how many abusable drug naive healthy subjects would require treatment to cause moderate or severe sedation (or drowsiness) in one subject, where moderate to severe sedation or drowsiness is defined as a VAS score of > 50 mm on a 100 mm scale bounded on the left by "no sedation or drowsiness” and on the right by "extreme sedation or drowsiness”; (ii) how many abusable drug naive healthy subjects would require treatment to cause moderate or severe nausea in one subject, where moderate to severe nausea is defined as a VAS score of > 50 mm on a 100 mm scale bounded on the left by "no nausea” and on the right by "extreme nausea”; (i
  • This questionnaire can be used to examine the overall drug effects of abusable drugs given intact and upon tampering, preferably in drug abusers and recreational drug users without the medical condition for which the drug is effective.
  • This questionnaire can be used to examine the overall drug liking of abusable drugs given intact and upon tampering, preferably in drug abusers and recreational drug users without the medical condition for which the drug is effective.
  • the "take again” questionnaire assesses whether subjects would take the abusable drug again if given the opportunity. The patient is asked "If given an opportunity, would you take this drug again? (circle one: YES or NO). This questionnaire can be used to examine the overall desirability of the drug experience with the abusable drugs taken intact and taken after . tampering, preferably in drug abusers and recreational drug users without the medical condition for which the drug is effective.
  • VAS visual analog scale
  • Three performance tasks may be employed for measuring skills related to driving.
  • the "critical tracking task” measures the patient's ability to control a displayed error signal in a first-order compensatory tracking task.
  • the error is displayed as a horizontal deviation of a cursor from the midpoint on a horizontal, linear scale.
  • Compensatory joystick movements correct the error by returning the cursor to the midpoint.
  • the frequency at which the patient loses the control is the critical frequency.
  • the critical tracking task measures the psychomotor control during a closed loop operation. It is a laboratory analog to on-the-road tracking performance.
  • the "stop signal task” measures motor impulsivity, which is defined as the inability to inhibit an activated or pre-cued response leading to errors of commission. The task requires patients to make quick key responses to visual go signals, i.e.
  • the main dependent variable is the stop reaction time on stop signal trials that represents the estimated mean time required to inhibit a response.
  • the Tower of London is a decision-making task that measures executive function and planning.
  • the task consists of computer generated images of begin- and end-arrangements of three colored balls on three sticks.
  • the subject's task is to determine as quickly as possible, whether the end- arrangement can be accomplished by "moving" the balls in two to five steps from the beginning arrangement by pushing the corresponding number coded button.
  • the total number of correct decisions is the main performance measure.
  • the amount of abusable drug in the dosage form is about 0.01 ⁇ g to 1500 mg. In other more preferred embodiments, the amount of abusable drug in the dosage form is about 0.1 ⁇ g to 1000 mg or about 0.1 ⁇ g to 1500 mg.
  • the amount of abusable drug in the dosage form is about 0.01 ⁇ g to 750 mg, or about 0.01 ⁇ g to about 500 mg, or about 0.01 ⁇ g to about 250 mg, or about 0.1 ⁇ g to about 500 mg, or 0.1 ⁇ g to about 250, or about 0.1 ⁇ g to about 250 mg, or about 1 ⁇ g to about 1500 mg, or 1 ⁇ g to about 1000 mg, or about 1 ⁇ g to about 100 mg, or about 5 ⁇ g to about 1500 mg, or about 5 ⁇ g to about 1000 mg, or about 5 ⁇ g to about 500 mg, or about 10 ⁇ g to about 1000 mg, or about 10 ⁇ g to about 500 mg, or about 100 ⁇ g to about 1000 mg, or about 1 mg to about 1500 mg, or about 1 mg to about 1000 mg, or about 1 mg ⁇ g to about 800 mg, or about 1 mg to about 500 mg.
  • the amount of ADER in the claimed composition may be about 1 mg to 1500 mg. In a preferred embodiment, the amount of ADER in the claimed composition may be about 10 mg to 800 mg. In a most preferred embodiment, the amount of ADER in the claimed composition may be about 50 mg to 600 mg.
  • the ratio of the abusable drug and the ADER is about 1:10,000 to about 10,000:1 by weight, preferably about 1:1000 to about 1000:1 by weight, more preferably 1 :250 to 250:1.
  • pH-dependent for purposes of the present invention is defined as having characteristics (e.g., dissolution) which vary according to environmental pH.
  • pH-independent for purposes of the present invention is defined as having characteristics (e.g., dissolution) which are substantially unaffected by pH.
  • bioavailability is defined for purposes of the present invention as the rate and extent to which the drug (e.g., the abusable drug) is absorbed from the unit dosage forms.
  • oral refers to any method of administration involving contact with the mouth and oral mucosa, including the ingestion of intact drugs (e.g., capsules, tablets, liquids swallowed whole), lingual, sublingual administration, buccal administration and transmucosal administration.
  • intact drugs e.g., capsules, tablets, liquids swallowed whole
  • lingual, sublingual administration e.g., buccal administration
  • transmucosal administration e.g.
  • AU oral pharmaceutical dosage forms of the invention are contemplated, including oral suspensions, tablets, capsules, lozenges, effervescent tablets, effervescent powders, powders, solutions, powders for reconstitution, transmucosal films, buccal products, oral mucoretentive products, oral gastroretentive tablets and capsules, orally disintegrating tablets, fast dissolving tablets, fast dispersing tablets, fast disintegrating dosage forms, administered as immediate release, delayed release, modified release, enteric coated, sustained release, controlled release, pulsatile release and extended release dosage form.
  • controlled release is interchangeable with “extended release”, “sustained release”, “modified release”, “delayed release” and the like. Such products provide a longer duration of action than conventional immediate release formulations of the same drugs and are usually administered every 8, 12 or 24 hours.
  • Controlled release dosage forms of the present invention release abusable drug from the oral dosage form at slower rate than immediate release formulations.
  • controlled release dosage forms release abusable drug at such a rate that blood (e.g., plasma) concentrations (levels) or therapeutic effects are maintained within the therapeutic range (above the minimum effective therapeutic concentration) but below toxic levels for intended duration (e.g., over a period of 1 to 24 hours, preferably over a period of time indicative of Q4, Q6, QS, Q12 or Q24H administration).
  • the controlled release formulations of the present invention provide therapeutic effects for a duration that is longer or substantially longer than the duration of meaningful or detectable plasma concentrations of abusable drug.
  • Controlled release dosage forms may be administered around the clock on a scheduled or time contingent basis, or on an as needed or PRN basis, e.g., Q3 PRN, Q4 PRN, Q6 PRN, Q8 PRN, Q12 PRN or Q24H PRN administration.
  • immediate release abusable drug for purposes of the present invention is abusable drug for oral administration in a dosage form which formulated to release the active drug from the dosage form immediately (i.e., without an attempt to delay or prolong the release of the active drug from the dosage form as is the case for extended release dosage forms).
  • an available parenteral formulation of abusable drug or a salt thereof may be used orally or a solution of abusable drug or a salt thereof may be prepared for the purpose of in vivo testing requiring immediate release abusable drug.
  • the controlled release formulations disclosed herein and the immediate release control formulations are dose proportional.
  • the pharmacokinetic parameters e.g., AUC and C max
  • the pharmacokinetic parameters of a particular dose can be inferred from the parameters of a different dose of the same formulation.
  • agonist means a ligand that binds to a receptor and alters the receptor state resulting in a biological response. Conventional agonists increase receptor activity, whereas inverse agonists reduce it (See Neubig et al, IUPHAR Committee on Receptor Nomenclature and Classification, Pharmacol Rev, 2003; Howlett et al., MoI Pharmacol, 1988).
  • opioid agonist means a molecule that causes a specific physiologic, pathophysiologic or pharmacologic effect after binding to an opioid receptor.
  • an "antagonist” is a drug or ligand that reduces the action of another drug or ligand, generally an agonist. Many antagonists act at the same receptor macromolecule as the agonist. (See Neubig et al, IUPHAR Committee on Receptor Nomenclature and Classification, Pharmacol Rev, 2003; Howlett et al., MoI Pharmacol, 1988).
  • receptor means a molecule within a cell, on a cell surface, on a membrane, in tissue, in fluid or otherwise found in humans that serve as a recognition or binding site to cause specific physiologic, pathophysiologic or pharmacologic effects.
  • the term “receptor” also means a cellular macromolecule, or an assembly of macromolecules, that is concerned directly and specifically in chemical signaling between and within cells. Combination of a hormone, neurotransmitter, drug, ligand, or intracellular messenger with its receptor(s) initiates a change in cell function (Neubig et al, IUPHAR Committee on Receptor Nomenclature and Classification, Pharmacol Rev, 2003).
  • benzodiazepine abuse in the context of the present invention includes intermittent use, recreational use and chronic use of abusable drugs alone or in conjunction with other drugs: (i) in quantities or by methods and routes of administration that do not conform to standard medical practice; (ii) outside the scope of specific instructions for use provided by a qualified medical professional; (iii) outside the supervision of a qualified medical professional; (iv) outside the approved instructions on proper use provided by the drug's legal manufacturer; (v) which is not in specifically approved dosage forms for medical use as pharmaceutical agents; (vi) where there is an intense desire for and efforts to procure same; (vii) with evidence of compulsive use; (viii) through acquisition by manipulation of the medical system, including falsification of medical history, symptom intensity, disease severity, patient identity, doctor shopping, prescription forgeries; (ix) where there is impaired control over use; (x) despite harm; (xi) by procurement from non
  • abusable drugs are: (i) substances which have medical applications for the prevention or treatment of diseases and disorders (i.e., "therapeutically active agents”); and (ii) substances which have the potential for being abused by drug abusers, recreational drug users and individuals with an addiction disorder for intermittent use, recreational use or chronic use, wherein the abusable drug is being abused for one or more of the following effects: mood alterations; euphoria, pleasure; a feeling of high; a feeling of drug liking; anxiolysis; mental stimulation; increased mental arousal; sedation; calmness; a state of relaxation; psychotomimesis; hallucinations; alterations in perception, cognition and mental focus; insomnia; hypersomnia; increased wakefulness or alertness; memory improvement; increased sexual gratification; increased sexual arousal; increased sexual desire and sexual anticipation; increased socialization; reduced
  • abusable drugs are, without limitation amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, nicotine, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof, but specifically excluding all opioid agonists.
  • abusable drug refers only to methylphenidate, or only to amphetamine analogs, or only to amphetamines and amphetamine analogs, or only to modafinil, or only to CNS or psychostimulants, or only to cannabinoid agonists, or only to nicotine, or only to alkylxanthines, or only to anorectics, or only to barbiturates, or only to benzodiazepine agonists, or only to non- benzodiazepine hypnotics and CNS depressants, or only to anxiolytic agents, or only to sedatives, or only to hypnotics, or only to controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended), or only to drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and their respective pharmaceutically acceptable salts, prodrugs or analogs in
  • abusable drug refers to one or more compounds selected from the group comprising methylphenidate, amphetamine analogs, amphetamines, modafinil, CNS or psychostimulants, cannabinoid agonists, nicotine, alkylxanthines, anorectics, barbiturates, benzodiazepine agonists, non- benzodiazepine hypnotics, CNS depressants, anxiolytics, sedatives, hypnotics, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended), and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and their respective pharmaceutically acceptable salts, prodrugs or analogs in racemic or enantiomeric form.
  • abusable drugs of the invention may be in unsalified form as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof.
  • abusable drugs of the invention may be pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymotphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof.
  • abusable drugs of the invention may be in unsalified form, pharmaceutically acceptable salts, prodrugs, esters or with a covalently bound pharmaceutically active moiety or mixtures thereof, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof.
  • a number of assays are available to determine whether a drug is a benzodiazepine agonist or benzodiazepine antagonist, using in vivo and in vitro bioassay systems.
  • Benzodiazepine agonist are known or readily determined by individuals who practice the art. All benzodiazepine agonists are contemplated by the invention, including benzodiazepine BZl (omega 1) receptor agonists.
  • the benzodiazepine agonist useful for the present invention may be selected from the group consisting of alprazolam, bromazepam, brotizolam, camazepam, chlordiazepoxide, cinolazepam, clobazam, clonazepam, clorazepate, desalkylflurazepam, diazepam, estazolam, flunitrazepam, flurazepam, halazepam, ketazolam, loprazolam, lorazepam, lormetazepam, medazepam, metaclazepam, midazolam, nitrazepam, nordazepam, oxaze
  • benzodiazepine agonist means a substance that binds to one or more benzodiazepine receptors or recognition sites, their subtypes and splice variants to exert an agonist or partial agonist effect.
  • the term "benzodiazepine receptor” or “benzodiazepine recognition site” includes one or more benzodiazepine receptors or recognition sites, the benzodiazepine BZl (omega 1) receptor, the gamma-aminobutyric acid (GABA)-benzodiazepine receptor complex, the gamma-aminobutyric acid type A (GABAA) receptor and their respective subtypes and splice variants.
  • GABA gamma-aminobutyric acid
  • GABAA gamma-aminobutyric acid type A
  • Non-benzodiazepine hypnotics and CNS depressants are known or readily determined by individuals who practice the art.
  • Non-benzodiazepine hypnotics and CNS depressants include sodium oxybate, diphenhydramine, chlorpheniramine, trazadone, amitriptyline, cyclobenzaprine, methocarbamol, carisoprodol, ramelteon, and drugs from the barbiturate, antihistamine, antidepressant and melatonin receptor agonist class.
  • Cannabinoid agonists are known or readily determined by individuals who practice the art.
  • the cannabinoid agonist useful for the present invention may be selected from the group consisting of inhibitors of cannabinoid agonist metabolism (e.g., without limitation, URB602, an inhibitor of monoacylglycerol lipase which catalyzes 2-arachidonoylglycerol hydrolysis) THC, nabilone, dronabinol, cannabidiol, 9-THC propyl analog, cannabidiol, cannabidiol propyl analog, carmabinol, cannabichromene, cannabichromene propyl analog, cannabigerol, cannabinoid terpenoids, cannabinoid flavonoids, endocannabinoids, anandamide, (R)- methanandamide,and 2-arachidonoylglycerol, THC-like ABC tricyclic cannabinoid
  • the cannabinoid agonist useful for the present invention may be selected from the group consisting of dexanabinol (HU211), BAY 38-7271 , Naphthalen- 1 -yl-(4-pentyloxynaphthalen- 1 -yl)methanone, THC (delta-9-tetrahydrocannabinol), nabilone, dronabinol, cannabidiol, cannabinol, cannabichromene, cannabigerol, cannabigerol, anandamide, (R)- methanandamide, 2-arachidonoylglycerol, HU210, desacetyllevonantxadol, CP55940, CP55244, URB602, or WIN55212-2 and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites
  • the cannabinoid agonist useful for the present invention may be selected from the group consisting of 9-THC propyl analog, endocannabinoids, cannabinoid terpenoids, cannabinoid flavonoids, inhibitors of cannabinoid agonist metabolism, inhibitors of monoacylglycerol lipase, cannabidiol propyl analogues, cannabichromene propyl analogues, THC-like ABC tricyclic cannabinoid analogues, synthetic AC bicyclic cannabinoid analogues, synthetic ACD tricyclic cannabinoid analogues, aminoalkylindole compounds or analogs of 2-Arylimino-5,6-dihydro-4H-l,3-thiazines and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an
  • the amount of the cannabinoid agonist used by the subject may be from about 10 ng to about 2000 mg, even up to about 2000 mg. More preferably, the amount of the cannabinoid agonist is from about 10 ng to about 1500 mg, even more preferably from about 0.1 mg to about 1000 mg, and most preferably, from about 0.1 mg to about 700 mg.
  • the cannabinoid agonist may be selected from compounds disclosed in U.S. Patent No. 7,217,732, 7,214,716, 7,169,942, 7,109,216, 7,091,216, 7,057,051, 6,995,184, 6,972,295, 6,943,266, 6,903,137, 6,864,291, 6,864,285, 6,525,087, 6,524,805, 6,509,367, 6,284,788, 5,948,777, 5,939,429, and 5,605,906, and in U.S. Patent Application No.
  • cannabinoid agonist shall include combinations of more than one cannabinoid agonist, and also include the unsalified agonist, mixed agonist-antagonists, partial agonists, pharmaceutically acceptable salts thereof, stereoisomers thereof, ethers and esters thereof, and mixtures thereof.
  • cannabinoid agonist for the purposes of the present invention, 1) drugs that enhance the effect of cannabinoid agonists by inhibiting their metabolism or reuptake (for example, anandamide amidase inhibitors) are considered to be cannabinoid agonists; 2) drugs that induce anandamide amidase inhibitor metabolism or induce CB], CB 2 and non-CBi/non-CB 2 cannabinoid agonist metabolism or enhance reuptake will be considered cannabinoid antagonists; 3) inverse cannabinoid agonists will be considered cannabinoid antagonists.
  • drugs that enhance the effect of cannabinoid agonists by inhibiting their metabolism or reuptake for example, anandamide amidase inhibitors
  • CB 2 and non-CBi/non-CB 2 cannabinoid agonist metabolism or enhance reuptake will be considered cannabinoid antagonists
  • a number of assays are available to determine whether a drug is a cannabinoid agonist, using in vivo and in vitro bioassay systems (Howlett et al., MoI Pharmacol, 1988; International Union of Pharmacology [IUPHAR], http://www.iuphar.org/index.html; Subcommittees on Cannabinoid Receptors The International Committee of Pharmacology Committee on Receptor Nomenclature and Classification [NC-IUPHAR], http ://www. iuphar . org/nciuphar .html) .
  • drugs that enhance the effect of cannabinoid agonists by inhibiting their metabolism or reuptake are also considered to be cannabinoid agonists.
  • cannabinoid agonist means a substance that binds to one or more cannabinoid receptor to exert an agonist or partial agonist effect.
  • cannabinoid receptor means a molecule that causes a specific physiologic, pathophysiologic or pharmacologic effect after binding to CBi, CB 2 , non-CBj/CB 2 cannabinoid sites, TRPVi receptors, as well as other G protein-coupled receptors (GPCRs) that form part of the endocannabinoid system (Wiley and Martin, Chemistry Physics of Lipids, 2002; Begg et al., Pharmacol Ther, 2005; Howlett et al., Neuropharmacol, 2004; Pertwee, AAPS Journal, 2005; International Union of Pharmacology (IUPHAR) Receptor Database; Howlett et al., MoI Pharmacol, 1988; International Union of Pharmacology [IUPHAR], http://www.iuphar.org/index.html; Subcommittees on Cannabinoid Receptors The International Committee of Pharmacology Committee on Receptor Nomenclature and Classification [
  • opioid receptor includes mu ( ⁇ ), delta ( ⁇ ), kappa (K) and/or nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptors, their subtypes and splice variants such as ⁇ 1 ; ⁇ 2 , ⁇ i, 6 2 , Ki, K 2 and K 3 , etc, regardless of whether they also bind to or influence other receptor systems (e.g., norepinephrine reuptake inhibition, serotonin reuptake inhibition, NMDA receptor antagonism).
  • NOP nociceptin/orphanin FQ peptide
  • Opioid antagonists are known or readily determined by individuals who practice the art.
  • the opioid antagonists useful for the present invention may be selected from the group consisting of naltrexone, methylnaltrexone, nalbuphine, naloxone, nalmefene, cyclazocine, cyclorphan, oxilorphan nalorphine, nalorphine dinicotinate, nalmefene, nadide and levallorphan.
  • Abusable drugs of the invention include stimulants, CNS-stimulants, psychostimulants, alkylxanthine, and anorectics, including, without limitation, compounds selected from the group comprising adrafanil, alkyxanthine derivatives, almitrine, amfetaminil, aminophylline, amiphenazole, ammonium camphocarbonate, amphetamine, bamifylline, benzfetamine, brolamfetamine, caffeine, cathine [(+)-norpseudoephedrine], cathinone, celastrin, chlo ⁇ hentermine, clonobenzorex, cropropamide, crotetamide, deanol, dextroamphetamine, diethylaminoethanol, diethylpropion, dimfline, doxapram, doxofylline, diprophylline, dyphylline, etamivan, etofylline, enp
  • the present invention anticipates the use of more than one abusable drug in some embodiments, given in the same formulation or in a different formulation, for use to treat, prevent or ameliorate the same disease or a different disease.
  • the invention allows for the use of lower doses of abusable drug by virtue of the inclusion or co-administration of an additional drug for the prevention or treatment of the sam medical condition.
  • an additional drug for the prevention or treatment of the sam medical condition.
  • abusable drug means an the abusable drug in unsalified form, a pharmaceutically acceptable salt, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof.
  • ADER includes an ADER compound as well as a mixture of two or more different ADER compounds
  • cannabinoid agonists includes a cannabinoid agonist as well as two or more different cannabinoid agonists in combination, and the like.
  • analgesic includes pharmacologic agents intended for or effective in the prevention and/or treatment of pain.
  • analgesics in addition to preventing or treating pain, provide salutary effects on signs and symptoms associated with pain.
  • analgesics in addition to relieving pain in patients with osteoarthritis, relieve stiffness, improve physical function, sleep and quality of life.
  • analgesics in addition to relieving pain in patients with neuropathic pain, reduce disability.
  • peripheral neuropathic pain e.g., acute and chronic inflammatory demeyelinating polyradiculopathy, alcoholic polyneuropathy, chemotherapy-induced polyneuropathy, complex regional pain syndrome (CRPS) Type I and Type II, entrapment neuropathies (e.g., carpal tunnel syndrome), HIV sensory neuropathy, iatrogenic neuralgias (e.g., postthoracotomy pain, postmastectomy pain), idiopathic sensory neuropathy, painful diabetic neuropathy, phantom limb pain, postherpetic neuralgia, trigeminal neuralgia, radiculopathy (e.g., cervical thoracic, lumbosacral), sciatica, acute herpes zoster pain, temporomandibular joint ⁇ disorder pain and postradiation plexopathy; and (ii) central neuropathic pain, e.g., compressive myel
  • acute pain refers to self-limiting pain that subsides over time and usually lasting less that about 30 days and more preferably lasting less than about 21 days. Acute pain does not include chronic conditions such as chronic neuropathy, chronic neuropathic pain and chronic cancer and non-cancer pain.
  • neuropathic pain is pain initiated or caused by a primary lesion or dysfunction of the nervous system and includes (i) peripheral neuropathic pain and (ii) central neuropathic pain.
  • chronic pain includes all non-neuropathic pain lasting more than 30 days, including inflammatory pain, noninflammatory pain, muscle pain, joint pain, fascia pain, visceral pain, bone pain and idiopathic pain.
  • ADHD is disorder as described in the Diagnostic and Statistical Manual of Mental Disorders (DSM), 1994, as revised in 2000 (DSM-IV-TR). ADHD is generally considered to be a developmental disorder, largely or entirely neurological in nature effecting 3-5 percent of the population characterized by a persistent pattern of inattention and/or hyperactivity-impulsivity, including hyperactivity, forgetfulness, poor impulse control, inattention, difficulty getting work done, procrastination, or organization problems and distractibility.
  • neurode disorders are disorders that affect the central nervous system (brain and spinal cord), the peripheral nervous system (peripheral nerves - cranial nerves included), or the autonomic nervous system (parts of which are located in both central and peripheral nervous system). See Adams & Victor's Principles of Neurology (McGraw-Hill Professional; 7 edition, 2000); Merritt's Textbook of Neurology (9th ed. Edited by Lewis P. Rowland. Baltimore: Williams and Wilkins, 1995); and Guide to Clinical Neurology (Mohr and Gautier, eds, New York, Churchill Livingstone, 1995).
  • psychiatric disorders and “mental illness” are used interchangeably.
  • a mental illness is an abnormal mental condition or disorder expressing symptoms that cause significant distress and/or dysfunction. This can involve cognitive, emotional, behavioral and interpersonal impairments.
  • psychiatric disorders are disorders described in the Diagnostic and Statistical Manual of Mental Disorders (DSM), 1994, as revised in 2000 (DSM-IV-TR).
  • analgesic effectiveness is defined for purposes of the present invention as a satisfactory prevention, reduction in or elimination of pain, along with a tolerable level of side effects, as determined by the human patient.
  • terapéuticaally-effective refers to the amount of an active agent sufficient to induce a desired biological result. That result may be alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • the term "effective amount” means the quantity of a compound according to the invention necessary to prevent, to cure, or at least partially arrest a sign or symptom for which the compound (e.g., abusable drug) has been prescribed to a subject.
  • abuse resistant and “abuse deterrent” are used interchangeably and include pharmaceutical compositions and methods to resist intentional, unintentional or accidental physical, mechanical, chemical or thermal manipulation or tampering of the dosage form (e.g., crushing, shearing, grinding, pulverizing, chewing, dissolving, melting, needle aspiration, syringe aspiration, syringe injection, solvent extraction, inhalation, insufflation, extraction by mechanical, thermal and chemical means, and/or filtration).
  • intentional, unintentional or accidental physical, mechanical, chemical or thermal manipulation or tampering of the dosage form e.g., crushing, shearing, grinding, pulverizing, chewing, dissolving, melting, needle aspiration, syringe aspiration, syringe injection, solvent extraction, inhalation, insufflation, extraction by mechanical, thermal and chemical means, and/or filtration).
  • abuse resistant and “abuse deterrent” also includes pharmaceutical compositions and methods to resist intentional, unintentional or accidental use or misuse of the dosage form: (i) in quantities or by methods and routes of administration that do not conform to standard medical practice; (ii) outside the scope of specific instructions for use provided by a qualified medical professional; (iii) outside the supervision of a qualified medical professional; (iv) outside the approved instructions on proper use provided by the drug's legal manufacturer; (v) in unapproved dosage forms; (vi) for compulsive use; (vii) through acquisition by manipulation of the medical system; (viii) for medically unapproved or unintended mood altering purposes.
  • pharmaceutical agent pharmaceutical agent
  • pharmaceutical agent pharmaceutical agent
  • active agent active agent
  • subject for purposes of treatment is used interchangeably with “patient”, “male”, “female”, and includes any human subject.
  • bioequivalent and “bioequivalence” means that the
  • excipient refers to a substance which does not interfere with the effectiveness or the biological activity of the active ingredients and which is not toxic to the subject.
  • pharmaceutically or therapeutically acceptable excipients or carriers may play a role in imparting or optimizing the rate and extent of absorption of the abusable drug or additional drugs in the pharmaceutical composition.
  • pharmaceutically or therapeutically acceptable excipients or carriers may play a role in stabilizing the abusable drug or additional drugs in the pharmaceutical composition.
  • the dosage form may include, in addition to abusable drug or a pharmaceutically acceptable salt thereof and ADER 5 other abuse deterrent or abuse resistant substances, process or technologies known in the art, including one or more aversive agents.
  • aversive agents include, without limitation, antagonists of abusable drugs, laxatives, cutaneous vasodilators, headache producing agents, emetics, emetogenic compound, nausea producing compounds, bittering agents, drugs that cause burning on irritation when in contact with tissue or mucous membranes (e.g., naso- mucosal irritants, oro-mucosal irritants, respiratory irritants), tissue irritants, gastrointestinal irritants, drugs that precipitate withdrawal effects, tissue dyes, lakes and colorants, beverage dyes, lakes and colorants, non-tissue staining beverage dyes, lakes and colorants (i.e, that do not stain or discolor the skin upon ingestion), fecal discolorants, urine discolorants, malodorous agents, opioid antagonists, benzodiazepine antagonists (e.g., flumazenil), cannabinoid antagonists and pharmacologic antagonists to co-abused drugs not contained
  • Such aversive agents may be in the dosage form in a releasable, partially releasable or a non-releasable form (i.e., sequestered), the latter being released on tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like). Further, in some embodiments, such aversive agents may be in the dosage form in an amount that does not produce an aversive effect or aversion in any, many or substantially all patients when taken in accordance with the prescribing information or the manufacturer's instructions (for example, in small quantities), but which produce an aversive effect when taken in excess (e.g., higher dose or more frequently).
  • one or more aversive agents may be added to the formulation in an aversive agent amount of less than about 80% by weight, preferably less than about 60% by weight, more preferably less than about 40% by weight of the dosage form, even more preferably less than about 20% by weight of the dosage form, and most preferably less than about 10 by weight of the dosage form (e.g., 0.000000000000001% to 1%, or 0.000000001% to 3%, or 0.0001% to 10%, or 0.001% to 5%, or 1% to 10%, or 0.001% to 2%, or 1% or 10%, or 2% to 7%) depending on the particular aversive agent used.
  • an aversive agent amount of less than about 80% by weight, preferably less than about 60% by weight, more preferably less than about 40% by weight of the dosage form, even more preferably less than about 20% by weight of the dosage form, and most preferably less than about 10 by weight of the dosage form (e.g., 0.000000000000001% to 1%, or 0.000000001%
  • the aversive agent in the dosage form may be about 0.00000000001 mg to about 2000 mg, or about 0.0000001 mg to about 1500 mg, or about 0.000001 mg to about 1000 mg, or about 0.0001 mg to about 1000 mg, or about 0.001 mg to about 1000 mg, or about 0.01 mg to about 1000 mg, or about 0.1 rag to about 1500 mg, or 1 mg to about 800 mg, or about 1 mg to about 500 mg, or about 1 mg to about 300 mg, or about 1 mg to about 150 mg, or about 5 mg to about 400 mg, or about 5 mg to about 200 mg, or about 0.00000000001 mg to about 200 mg, or about 0.00000000001 mg to about 100 mg, or about 0.00000000001 mg to about 50 mg, or about 0.0000001 mg to about 200 mg, or about 0.0000001 mg to about 100 mg, or about 0.00001 mg to about 400 mg, or about 0.0001 mg to about 300 mg.
  • the present invention can include one or more aversive agents, selected from the group including, without limitation antagonists of abusable drugs, laxatives, cutaneous vasodilators headache producing agents, emetics, emetogenic compound, nausea producing compounds, bittering agents, drugs that cause burning on irritation when in contact with tissue or mucous membranes (e.g., naso-mucosal irritants, oro- mucosal irritants, respiratory irritants), tissue irritants, gastrointestinal irritants, drugs that precipitate withdrawal effects, tissue dyes, lakes and colorants, beverage dyes, lakes and colorants, non-tissue staining beverage dyes, lakes and colorants, fecal discolorants, urine discolorants, malodorous agents, opioid antagonists, benzodiazepine antagonists, cannabinoid antagonists, and pharmacologic antagonists to co-abused drugs not contained in the dosage form.
  • aversive agents selected from the group including, without limitation antagonists of
  • the aversive agent is a pharmaceutically acceptable agent that produces an aversive effect only when the dosage form of the invention containing the aversive agent is abused, for example, when taken in excess of medically approved doses, taken in excess of approved doses in the manufacturer's prescribing information, taken after tampering of the dosage form (e.g., mechanical, thermal, chemical, solvent tampering), ingestion in ways not medically recommended, administration by routes not approved for the dosage form (e.g., intranasal, inhalation, intravenous) or in a manner inconsistent with the manufacturer's prescribing information.
  • tampering of the dosage form e.g., mechanical, thermal, chemical, solvent tampering
  • routes not approved for the dosage form e.g., intranasal, inhalation, intravenous
  • the amount of aversive agent in the dosage form of the present invention can be a fixed ratio in relation to the amount of abusable drug in the dosage form.
  • aversive effects can be avoided under conditions of proper medical use (e.g., manufacturers prescribing directions).
  • the quantity of aversive agent consumed will exceed the "no effect” or "minimum effect” threshold, thereby producing one or more aversive effects, for example, e.g., nausea, emesis, diarrhea, laxation, cutaneous vasodilation, headache, bitter taste, naso-mucosal irritation, oro- mucosal irritation, precipitation of abstinence from the abusable drug of the dosage form, precipitation of abstinence from a co-abused drug which is not part of the dosage form, reduction of the pleasurable, mood altering, rewarding, reinforcing, stimulant, depressant or other psychic and physiologic effects of the abusable drug or a co-abused drug, etc.).
  • aversive effects for example, e.g., nausea, emesis, diarrhea, laxation, cutaneous vasodilation, headache, bitter taste, naso-mucosal irritation, oro- mucosal irritation, precipit
  • the "no effect" or “minimum effect” threshold amount of aversive agent can be exceeded when the dosage form of the invention is taken in excess of the manufacturer's recommendation by a factor of about 1.5, or about 2, or about 2.5, or about 3, or about 4, or about 5, or about 6, or about 7, or about 8, or about 10, or more than 10.
  • the production of an aversive effect can reduce or stop further abuse of the dosage form, thereby reducing the harm or toxicity of the drug in the subject who is tampering, misusing or abusing the dosage form, e.g., addicts, drug abusers and recreational drug users.
  • bittering agents can be employed including, for example and without limitation, T2R or TAS2R receptor agonists, phenylthiourea (phenylthiocarbamide), natural, artificial and synthetic flavor oils and flavoring aromatics and/or oils, oleoresins and extracts derived from plants, leaves, flowers, fruits, and so forth, and combinations thereof.
  • Nonlimiting representative flavor oils include spearmint oil, peppermint oil, eucalyptus oil, oil of nutmeg, allspice, mace, oil of bitter almonds, menthol and the like.
  • Also useful bittering agents are artificial, natural and synthetic fruit flavors such as citrus oils including lemon, orange, lime, grapefruit, and fruit essences and so forth.
  • Additional bittering agents include sucrose derivatives (e.g., sucrose octaacetate), chlorosucrose derivatives, quinine and quinine salts, quinidine and quindine salts and the like.
  • the preferred bittering agent for use in the present invention is denatonium, denatonium benzoate and denatonium saccharide.
  • a dosage form including a bittering agent preferably discourages improper usage of the tampered dosage form by imparting a disagreeable taste to the tampered dosage form.
  • the aversive agent in the dosage form may be denatonium, denatonium saccharide or denatonium benzoate, in a quantity expressed as mg of denatonium, of about 0.00000001 nag to about 100 mg, or about 0.000001 mg to about 100 mg, or about 0.0001 mg to about 100 mg, or about 0.0001 mg to about 20 mg, or about 0.0001 mg to about 10 mg, or about 0.0001 mg to about 5 mg, or about 0.0001 mg to about 2 mg, or about 0.0001 tng to about 1 mg, about 0.0001 mg to about 50 mg, or about 0.00000001 mg to about 50 mg, or about 0.00000001 mg to about 20 mg, or about 0.01 mg to about 20 mg, or about 0.01 mg to about 10 mg, or about 0.01 mg to about 5 mg, or about 0.01 mg to about 2 mg, or about 0.01 mg to about 1 mg, or about 0.01 mg to about 1 mg, or about 0.01 mg to about 1 mg, or about 0.01 mg to about 1 mg, or about
  • the aversive agent in the dosage form may be quinine or a pharmaceutically acceptable salt of quinine, in a quantity expressed as mg of quinine, of about 0.00001 mg to about 300 mg, or about 0.00001 mg to about 200 mg, or about 0.00001 mg to about 100 mg, or about 0.00001 mg to about 75 mg, or about 0.00001 mg to about 50 mg, or about 0.00001 mg to about 25 mg, or about 0.00001 mg to about 20 mg, or about 0.00001 mg to about 10 mg, or about 0.00001 mg to about 5 mg, or about 0.00001 mg to about 2.5 mg, or about 0.00001 mg to about 1 mg, or about 0.001 mg to about 300 mg, or about 0.001 mg to about 200 mg, or about 0.001 mg to about 100 mg, or about 0.001 mg to about 75 mg, or about 0.001 mg to about 50 mg, .
  • 0.001 mg to about 25 mg or about 0.001 mg to about 20 mg, or about 0.001 mg to about 10 mg, or about 0.001 mg to about 5 mg, or about 0.001 mg to about 2.5 mg, or about 0.001 mg to about 1 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 100 mg, or about 1 mg to about 75 mg, or about 1 mg to about 50 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 1 mg to about 2.5 mg.
  • emetic agents can be employed including, for example and without limitation, zinc and pharmaceutically acceptable salts thereof (e.g., zinc oxide, zinc gluconate, zinc acetate, zinc sulfate, zinc carbonate), dopamine agonists, apomorphine, ipecac, ipecacuanha, emetine, emetine (methylcephaeline), cephaeline, psychotrine, O-methylpsychotrine, ammonium chloride, potassium chloride, magnesium sulfate, ferrous gluconate, ferrous sulfate, aloin, algarot or antimonious oxychloride, antimony trichloride, folate, folic acid, niacin (niacin) and nicotinamide.
  • zinc and pharmaceutically acceptable salts thereof e.g., zinc oxide, zinc gluconate, zinc acetate, zinc sulfate, zinc carbonate
  • dopamine agonists e.g., zinc
  • the aversive agent in the dosage form may be zinc in the form of elemental zinc or a pharmaceutically acceptable salt of zinc, in a quantity expressed as mg of elemental zinc, of about 1 mg to about 400 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 150 mg, or about 1 mg to about 100 mg, or about 1 mg to about 90 mg, or about 1 mg to about 80 mg, or about 1 mg to about 70 mg, or about 1 mg to about 60 mg, or about 1 mg to about 50 mg, or about 1 mg to about 45 mg, or about 1 mg to about 40 mg, or about 1 mg to about 40 mg, or about 1 mg to about 40 mg, or about 1 mg to about 35 mg, or about 1 mg to about 30 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 5 mg to about 400 mg, or about 5 mg to about 300 mg, or about 5 mg to about 200 mg, or about 5 mg to
  • Various irritants can be employed including, for example and without limitation transient receptor potential vanilloid 1 (TRPVl or VRl) agonists (including resiniferanoids, capsaicinoids, phorboid vanilloids, and terpenoid 1,4-unsaturated dialdehydes, capsaicin, capsaicin analogs and derivatives, resiniferatoxin, olvanil, pipeline, zingerone, anandamide, 12- and 15-(S)- hydroperoxy-eicosatetraenoic acids, 5 and 15-(S)-hydroxyeicosatetraenoic acids, phorbol 12-phenylacetate 13-acetate 20-homovanillate, 2 phorbol 12,13- didecanoate 20-homovanillate, leukotriene B(4), tinyatoxin, heptanoylisobutylamide, N-(3 -acyloxy-2-benzylpropy 1 )-N'- di
  • Various cutaneous vasodilators can be employed including, for example and without limitation, niacin acid, nicotinuric acid, beta- hydroxybutyrate and nicotinic receptor (e.g., HM74A or GPRl 09A) agonists.
  • niacin acid nicotinuric acid
  • beta- hydroxybutyrate beta- hydroxybutyrate
  • nicotinic receptor e.g., HM74A or GPRl 09A
  • the aversive agent in the dosage form may be niacin, in a quantity of about 1 nig to about 400 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 150 mg, or about 1 mg to about 100 mg, or about 1 mg to about 90 mg, or about 1 mg to about 80 mg, or about 1 mg to about 70 mg, or about 1 mg to about 60 mg, or about 1 mg to about 50 mg, or about 1 mg to about 45 mg, or about 1 mg to about 40 mg, or about 1 mg to about 40 mg, or about 1 mg to about 40 mg, or about 1 mg to about 35 mg, or about 1 mg to about 30 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 5 mg to about 400 mg, or about 5 mg to about 300 mg, or about 5 mg to about 200 mg, or about 5 mg to about 150 mg, or about 5 mg to about 100 mg, or about 10 mg to about
  • tissue dyes, lakes and colorants, beverage dyes, lakes and colorants, non-tissue staining beverage dyes, lakes and colorants, fecal discolorants, urine discolorants can be employed including, for example and without limitation, Curcumin, Riboflavin, Tartrazine, Quinoline yellow, Sunset yellow FCF, Carmine, Carmoisine, Amaranth, Ponceau 4R, Erythrosine, Allura red AC, Patent blue V, Indigo carmine, Brilliant blue FCF, Chlorophylls, Copper complexes of chlorophylls and chlorophyllins, Green S, i Caramel, Brilliant black BN, Vegetable carbon, Carotenoids, Alpha-, beta-, gamma-carotene, Capsanthin, Capsorubin, Lycopene, Beta-apo-8' carotenal, Ethyl ester of beta-apo-8' carotenoic acid, Xanthophylls, Lutein, Can
  • dyes used interchangeably and refer to one or more pharmaceutically acceptable dyes, lakes or colorants which may be: (i) tissue staining; (ii) non-tissue staining; (iii) beverage staining; (iv) urine discolorant; and/or (v) fecal discolorant.
  • Various laxatives can be employed including, for example and without limitation, Bis(p-hydroxyphenyl)pyridyl-2-rnethane, Bisacodyl, bisoxatin, anthraquinone, anthraquinone analogs and derivatives (e.g., buckthorn, casanthranol, cascara, hydroxy anthracene, glucofrangulin ), dantron, danthron, docusate (e.g., docusate sodium, docusate calcium, docusate potassium), gastrointestinal chloride channel activators (e.g., chloride channel subtype 2 activators), lubiprostone, magenesium salts (e.g., magnesium citrate, magnesium hydroxide, magnesium oxide), mannitol, oxyphenisatine, polyethylene glycol, poly(ethylene oxide) [PEO-1500], sodium phosphate, phenolphthalein, senna, senna constituents
  • the aversive agent in the dosage form may be a laxative in the amount of about 0.001 mg to about 300 mg, or about 0.001 mg to about 200 mg, or about 0.001 mg to about 100 mg, or about 0.001 mg to about 75 mg, or about 0.001 mg to about 50 mg, or about 0.001 mg to about 25 mg, or about 0.001 mg to about 20 mg, or about 0.001 mg to about 10 mg, or about 0.001 mg to about 5 mg, or about 0.001 mg to about 2.5 mg, or about 0.001 mg to about 1 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 100 mg, or about 1 mg to about 75 mg, or about 1 mg to about 50 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 1 mg to about 2.5 mg.
  • Aversive agents may include compounds found on the FDA EAFUS database (http://vm.cfsan.fda.gov/ ⁇ dms/eafus.html); FDA Food Additives Status List (http://www.cfsan.fda.gov/ ⁇ dms/opa-appa.html); FDAGRAS list and database; FDA Color Additive Status List
  • aversive agents may, in some embodiments be used in the dosage form of the invention for purposes other than as aversive agents, or for both aversive and non-aversive purposes.
  • non-aversive uses can include, without limitation, pharmaceutical purposes and pharmacologic purposes.
  • the laxative agent may be used to counteract the constipating effects of the abusable dosage form of the invention.
  • zinc and pharmaceutically acceptable salts of zinc and niacin may be used for pharmaceutical purposes (e.g., pharmaceutical optimization, drug release and drug stability).
  • the dosage form includes both an immediate release and extended release component.
  • the dosage form includes a capsule within a capsule, each capsule containing a different drug or the same drug intended for treating the same or a different medical condition.
  • the outer capsule may be an enteric coated capsule or a capsule containing an immediate release formulation to provide rapid plasma concentrations or a rapid onset of effect or a loading dose and the inner capsule contains an extended release formulation.
  • up to 3 capsules within a capsule are contemplated as part of the invention.
  • the dosage form involves one or more tablets within a capsule, wherein the abusable drug is either in the tablet and/or in one of the capsules.
  • the formulation is ingested orally as a tablet or capsule, preferably as a capsule.
  • the formulation is administered bucally.
  • the formulation is administered sublingually.
  • salts refers to a salt which is toxicologically safe for human and animal administration.
  • Nonlimiting examples of salts include hydrochlorides, hydrobromides, hydroiodides, sulfates, bisulfates, nitrates, citrates, tartrates, bitartrates, phosphates, malates, maleates, napsylates, fumarates, succinates, acetates, terephlhalates, pamoates and pectinates.
  • the abusable drug pharmaceutical composition is a salt or complex of inorganic cation salts, organic salts such primary, secondary, tertiary and quaternary amines include substituted amines
  • suitable pharmaceutically acceptable salts of abusable drugs include any of the inorganic cation salts such as sodium, potassium, lithium, magnesium, calcium, cesium, ammonia, ferrous, zinc, manganous, aluminum, ferric, and manganic; organic salts with primary, secondary, tertiary and quaternary amines, or mixtures thereof.
  • Examples of such primary, secondary, tertiary and quaternary amines include substituted amines including but not limited to naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and mixtures thereof. More specifically, suitable amines include but are not limited to tromethamine, triethylamine, tripropylamine, dropopizine, 2-dimethylaminoethanol, 2- diethylaminoethanol, lysine, arginine, ornithine, histidine, caffeine, procaine, N-ethylpiperidine, hydrabamine, choline, betai ⁇ e, ethylenediamine, glucosamine, fr ⁇ -(hydroxymethyl)aminomethane, N-methylglucamine, methylglyc amine, theobromine, piperazine, piperidine, polyamine resins and the like, and mixtures thereof.
  • suitable amines include but are not limited to tromethamine, triethyl
  • examples of suitable pharmaceutically acceptable salts of abusable drugs include aminoalcohols chosen from the group consisting of ethanolarnine, 3 -amino- 1 -prop anol, (i?)-l-amino-2- propanol, (S)-I -amino-2-propanol, 2-amino-l,3-propandiol, N-(2- hydroxyethyl)pyrrolidine, D-glucamine and L-prolin ⁇ l, D-glucosamine, and iV-methylglucosamine.
  • aminoalcohols chosen from the group consisting of ethanolarnine, 3 -amino- 1 -prop anol, (i?)-l-amino-2- propanol, (S)-I -amino-2-propanol, 2-amino-l,3-propandiol, N-(2- hydroxyethyl)pyrrolidine, D-glucamine and L-prolin ⁇ l
  • examples of suitable pharmaceutically acceptable salts of abusable drugs include alkali and alkaline earth metals and salts of an organic nature, such as the salts of basic amino acids.
  • the present invention may be used alone or in combination with other drugs to provide additive, complementary, or synergistic therapeutic effects or for the treatment of entirely different medical conditions.
  • Other pharmaceutically active ingredients from various therapeutic classes may also be used in combination with the present invention. They include, but are not limited to decongestants, analgesics, analgesic adjuvants, antidepressants, antipsychotics, anxiolytics, hypnotics, sedatives, anti-ADHD drugs, psychostimulants, drugs to treat urinary incontinence, antihistamines, expectorants, antitussives, diuretics, anti-inflammatory agents, antipyretics, antirheumatics, antioxidants, laxatives, local anesthetics, proton pump inhibitors, motility modifying agents, vasodilators, inotropes, beta blockers, beta adrenergic agonists, drugs to treat asthma and COPD, antiinfectives, antimigraine agents, antihypertensives, antianginal agents, gastric acid reducing agents, anti-ulcer agents, anticoagulants, lipid and cholesterol lowering drugs, anti-dia
  • pathological states are used interchangeably and are intended to have their broadest interpretation to refer to any physiologic, pathologic or pathophysiologic state in a human that can be prevented, treated, managed or altered to produce a desired, usually beneficial effect.
  • the oral abusable drug is intended to prevent or treat pain.
  • a co-administered drug in the same or different dosage form, by any route of administration may be used to provide additive, complementary, superadditive or synergistic therapeutic analgesic effects, including other NSAIDs, NO-NSAIDs, COX-2 selective inhibitors, acetaminophen, nitroparacetamol, nitric oxide donors, tramadol, beta adrenergic agonists, alpha-2 agonists, selective prostanoid receptor antagonists, NO-opioid receptor agonists, local anesthetics, purinergic P2 receptor antagonists, NMDA receptor antagonists, gabapentin, pregabalin, gabapentinoids, ligands of alpha(2)delta subunits of voltage-gated calcium channels, neuronal nicotinic receptor agonists, calcium channel antagonists, sodium channel blockers, superoxide dismutase
  • the ADER agent of the invention is selected from among hydrogenated Type I or Type II vegetable oils.
  • the ADER agent of the invention is selected from among polyoxyethylene stearates.
  • the ADER agent of the invention is selected from among polyoxyethylene distearates.
  • the ADER agent of the invention is selected from among polyoxyethylene stearates or distearates.
  • the ADER agent of the invention is selected from among hydrogenated Type I or Type II vegetable oils; said invention also including an aversive agent.
  • the ADER agent of the invention is selected from among polyoxyethylene stearates; said invention also including an aversive agent.
  • the ADER agent of the invention is selected from among polyoxyethylene distearates; said invention also including an aversive agent.
  • the ADER agent of the invention is selected from among polyoxyethylene stearates or distearates; said invention also including an aversive agent.
  • the ADER agent of the invention excludes hydrogenated Type I vegetable oils.
  • the ADER agent of the invention excludes hydrogenated Type II vegetable oils.
  • the ADER agent of the invention excludes hydrogenated Type I or Type II vegetable oils.
  • the ADER agent of the invention excludes polyoxyethylene stearates.
  • the ADER agent of the invention excludes polyoxyethylene distearates.
  • the dosage form includes two or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate;
  • the dosage form includes hydrogenated Type I or Type II vegetable oils in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
  • the dosage form includes polyoxyethylene stearates in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
  • the dosage form includes polyoxyethylene distearates in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
  • the dosage form includes glycerol monostearate in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
  • cumulative) amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
  • cumulative) amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
  • hydrogenated vegetable oils of the present invention include, without limitation, hydrogenated cottonseed oil (e.g., Akofine ® ; Lubritab ® ; Sterotex ® NF), hydrogenated palm oil (Dynasan ® P60; Softisan ® 154), hydrogenated soybean oil (Hydrocote ® ; Lipovol HS-K ® ; Sterotex ® HM) and hydrogenated palm kernel oil (e.g., Hydrokote ® 112).
  • polyoxyethylene stearates and distearates of the present invention include, without limitation, Polyoxyl 2, 4, 6, 8, 12, 20, 30, 40, 50, 100 and 150 stearates (e.g., Hodag ® DGS; PEG-2 stearate; Acconon ® 200-MS; Hodag ® 20-S; PEG-4 stearate; Cerasynt ® 616; Kessco ® PEG 300 Monostearate; Acconon ® 400-MS; Cerasynt ® 660; Cithrol ® 4MS; Hodag ® 60-S; Kessco ® PEG 600 Monostearate; Cerasynt ® 840; Hodag 100-S; Myrj ® 51; PEG-30 stearate; polyoxyethylene (30) stearate;Crodet ® S40; E431; Emerest ® 2672; Atlas G-2153; Crodet ® S50) and
  • waxes of the present invention include, without limitation: (i) animal waxes; (ii) insect waxes; (iii) vegetable waxes; (iv) mineral waxes; (v) petroleum waxes; (vi) synthetic waxes; (vi) nonionic emulsifying waxes or cetomacrogol emulsifying wax (e.g., Collone TV/TM; Crodex NTM; Emulgade 1000NITM; Permulgin DTM; PolawaxTM; Ritachol 2000; T- WaxTM); (vii) anionic emulsifying wax (e.g., Collone HVTM; Crodex ATM; Cyclonette wax; Lanette wax SXTMBP); (Vz ⁇ z ⁇ carnauba wax (also known as Brazil wax; caranda wax; E903); (ix) microcrystalline wax (also known as amorphous wax; E907; petroleum ceresin; petroleum wax (microcrystalline)); (x) yellow
  • the dosage form is devoid of animal waxes. In other embodiments, the dosage form is devoid of insect waxes. In other embodiments, the dosage form is devoid of vegetable waxes. In other • embodiments, the dosage form is devoid of mineral waxes. In other embodiments, the dosage form is devoid of petroleum waxes. In other embodiments, the dosage form is devoid of synthetic waxes. In other embodiments, the dosage form is devoid of nonionic emulsifying waxes or cetomacrogol emulsifying wax. hi other embodiments, the dosage form is devoid of anionic emulsifying wax. In other embodiments, the dosage form is devoid of carnauba wax.
  • the dosage form is devoid of microcrystalline wax. In other embodiments, the dosage form is devoid of yellow wax. In other embodiments, the dosage form is devoid of white wax. hi other embodiments, the dosage form is devoid of cetyl esters wax. In other embodiments, the dosage form is devoid of hydrogenated castor oil. In other embodiments, the dosage form is devoid of lanolin alcohols, hi other embodiments, the dosage form is devoid of lanolin, hi other embodiments, the dosage form is devoid of glyceryl palmitostearate. hi other embodiments, the dosage form is devoid of cetostearyl alcohol, hi other embodiments, the dosage form is devoid of beeswax.
  • the abusable drug is combined with beeswax, hydroxypropyl methyl cellulose (e.g, HPMC Kl 5M), silicon dioxide (alone or in combination with Al 2 O 3 ; e.g, Aerosil ® , Aerosil ® 200, Aerosil ® COK84).
  • beeswax hydroxypropyl methyl cellulose
  • HPMC Kl 5M hydroxypropyl methyl cellulose
  • silicon dioxide alone or in combination with Al 2 O 3 ; e.g, Aerosil ® , Aerosil ® 200, Aerosil ® COK84.
  • the abusable drug is combined with hydrogenated cottonseed oil (e.g., Sterotex ® NF), hydroxypropyl methyl cellulose (e.g, HPMC Kl 5M), fractionated coconut oil and silicon dioxide (alone or in combination with AI 2 O 3 ; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84).
  • hydrogenated cottonseed oil e.g., Sterotex ® NF
  • hydroxypropyl methyl cellulose e.g, HPMC Kl 5M
  • fractionated coconut oil and silicon dioxide alone or in combination with AI 2 O 3 ; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84.
  • the abusable drug is combined with glycerol monostearate (e.g., Cithrol ® GMS), hydroxypropyl methyl cellulose (e.g, HPMC KlOOM) and silicon dioxide (alone or in combination with Al 2 O 3 ; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84).
  • glycerol monostearate e.g., Cithrol ® GMS
  • HPMC KlOOM hydroxypropyl methyl cellulose
  • silicon dioxide alone or in combination with Al 2 O 3 ; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84.
  • the abusable drug is combined with hydrogenated palm kernel oil (e.g., Hydrokote ® 112), hydroxypropyl methyl cellulose (e.g, HPMC Kl 5M) and silicon dioxide (alone or in combination with Al 2 O 3 ; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84).
  • hydrogenated palm kernel oil e.g., Hydrokote ® 112
  • hydroxypropyl methyl cellulose e.g, HPMC Kl 5M
  • silicon dioxide alone or in combination with Al 2 O 3 ; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84.
  • release rate modifiers may be incorporated. Release rate modifiers can also have additional useful properties that optimize the formulation.
  • cellulose and cellulose derivatives including, without limitation cellulose acetate, microcrystalline cellulose, powdered cellulose, cellulose acetate phthalate, hydroxyethyl cellulose, silicified microcrystalline cellulose, hydroxypropyl cellulose, hydroxyethylmethyl cellulose, low-substituted hydroxypropyl cellulose, carboxymethylcellulose, carboxymethylcellulose calcium, hypromellose acetate succinate, hypromellose phthalate and ethylcellulose.
  • coconut oil products including without limitation, coconut oil, fractionated coconut oil, cetyl alcohol, lauric acid and medium chain triglycerides (e.g., Bergabest; caprylic/capric triglyceride; Captex 300; Captex 355; Crodamol GTC/C; glyceryl tricaprylate/caprate; Labrafac CC; MCT oil; Miglyol 810TM; Miglyol 812TM; Myritol; Neobee M5TM; NesatolTM; oleum neutrale; oleum vegetable whatsoever; thin vegetable oil; Waglinol 3/9280TM).
  • the coconut oil is fractionated coconut oil.
  • hydroxypropyl methyl cellulose e.g, HPMC Kl 5M
  • HPMC Kl 5M hydroxypropyl methyl cellulose
  • a variety of agents may be incorporated into the ADER invention as thixotropes (e.g., fumed silicon dioxides, Aerosil®, Aerosil® COK84, Aerosil® 200, etc.).
  • Thixotropes enhance the pharmaceutical formulations of the invention by increasing the viscosity of solutions during attempted extraction, complementing the action of HPMCs. They may also provide a tamper resistance by helping to retain the structure of dosage units that have been heated to temperatures greater than the melting point of the base excipient (Aerosils are unaffected by heat).
  • the present invention can include one or more
  • ADER agents Any amount of ADER may be used. In some embodiments, the total amount of ADER agent is about 5 to about 98 percent, preferably 7 to 90 percent and more preferably 10 to 85 percent on a dry weight basis of the composition.
  • the ADER can prevent less than or equal to about 98%, 90%, 80% 75%, 60%, 50%, 45%, 40%, 33%, 30%, 25%, 15%, 10%, 8%, 5%, or 2% of the total amount of drug in a dosage form from being recovered from a solvent in contact with a dosage form of the present invention.
  • the abuse deterrent, extended release, alcohol dose dumping protective and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a hydrogenated Type I vegetable oil.
  • the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a hydrogenated Type II vegetable oil.
  • the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a hydrogenated Type I or Type II vegetable oil.
  • the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a polyoxyethylene stearate. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a polyoxyethylene distearate. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a glycerol monostearate. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents,, each a wax.
  • the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) a hydrogenated Type I vegetable oil; (ii) a hydrogenated Type II vegetable oil; (iii) a polyoxyethylene stearate; (iv) a polyoxyethylene distearate; (v) a glycerol monostearate; and (vi) a wax.
  • the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) a hydrogenated Type I vegetable oil; (ii) a hydrogenated Type II vegetable oil; (iii) a polyoxyethylene stearate; (iv) a polyoxyethylene distearate; (v) a glycerol monostearate; and (vi) a wax, said dosage form having a melting point > 50°C, or > 55°C, or > 60 0 C, or > 65°C, or > 70 0 C, or > 75°C, or > 80 0 C, or > 85°C, or > 9O 0 C, or between 50 and 60 0 C, or between 55 and 65°C, or between 60 and 7O 0 C, or between 65 and 75°C, or between 70 and 80 0 C, or
  • the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) hydrogenated Type I or Type II vegetable oil; (ii) polyoxyethylene stearate or distearate; (iii) a glycerol monostearate; and (iv) a wax.
  • the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) hydrogenated Type I or Type II vegetable oil; (ii) polyoxyethylene stearate or distearate; (iii) a glycerol monostearate; and (iv) a wax, said dosage form having a melting point > 50 0 C, or > 55°C, or > 60 0 C, or > 65°C, or > 70 0 C, or > 75°C, or > 80 0 C, or > 85°C, or > 90 0 C, or between 50 and 60 0 C, or between 55 and 65°C, or between 60 and 70 0 C, or between 65 and 75°C, or between 70 and 80 0 C, or between 75 and 85°C S or between 80 and 9O 0 C, or between 90 and 100
  • the present invention can also optionally include other ingredients to enhance dosage form manufacture from a pharmaceutical composition of the present invention and/ or alter the release profile of a dosage form including a pharmaceutical composition of the present invention.
  • Some embodiments of the present invention include one or more pharmaceutically acceptable fillers, diluents, glidants and lubricants of various particle sizes and molecular weights.
  • the dosage form according to the invention may also comprise a coating which is resistant to gastric juices and dissolves as a function of the pH value of the release environment. By means of this coating, it is possible to ensure that, when correctly administered, the dosage form according to the invention passes through the stomach undissolved and the active ingredient is only released in the intestines.
  • the dosage form may include a surfactant ingredient to impart suitable formulation characteristics to the composition.
  • Surfactants may be hydrophilic preferably selected from the group consisting of non-ionic hydrophilic surfactants and anionic hydrophilic surfactants or the surfactant may have hydrophobic properties.
  • non-ionic hydrophilic surfactants are polyoxyethylene sorbitan esters, cremophores and poloxamers.
  • anionic surfactants are sodium lauryl sarcosinate, docusate and pharmaceutically acceptable docusate salts. Also a mixture of these surfactants can be used.
  • the formulation optionally comprises auxiliary materials.
  • auxiliary materials are (i) Binders such as acacia, alginic acid and salts thereof, cellulose derivatives, methylcellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, magnesium aluminum silicate, polyethylene glycol, gums, polysaccharide acids, bentonites, hydroxypropyl methylcellulose, gelatin, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer, crospovidone, povidone, polymethacrylates, hydroxypropylmethylcellulose, hydroxypropylcellulose, starch, pregelatinized starch, ethylcellulose, tragacanth, dextrin, microcrystalline cellulose, sucrose, or glucose, and the like; (ii) Disintegrants such as starches, pregelatinized corn starch, pregelatinized starch, celluloses, cross-linked
  • compositions and dosage form of the invention may further contain one or more pharmaceutically acceptable excipients.
  • pharmaceutically acceptable excipients can play a small or significant role in the behavior of the dosage form, depending on the choice of excipient, quantity of excipient and interaction with other constituents of the dosage form and the gastrointestinal tract.
  • excipients are well known in the art and include, without limitation, excipients referenced in the FDA EAFUS database (http://vmxfsan.fda.gov/ ⁇ dms/eafus.html); FDA Food Additives Status List (http://www.cfsan.fda.gov/ ⁇ dms/opa-appa.html); FDAGRAS list and database; FDA Color Additive Status List
  • the dosage form according to the invention may also comprise a coating which is resistant to gastric juices and dissolves as a function of the pH value of the release environment. [00582] By means of this coating, it is possible to ensure that, when correctly administered, the dosage form according to the invention passes through the stomach undissolved and the active ingredient is only released in the intestines. [00583] In one preferred embodiment of the invention, the formulation is ingested orally as a tablet or capsule, preferably as a capsule. In another preferred embodiment of the invention, the formulation is administered bucally. In yet another preferred embodiment of the invention, the formulation is administered sublingually.
  • the invention provides for methods and pharmaceutical compositions to prevent or minimizing excessive peak concentrations (dose dumping) of therapeutic doses of extended release abusable drugs for medical purposes, when they are co-ingested with alcohol.
  • the invention provides for methods and pharmaceutical compositions to achieve an extended release formulation of abusable drugs.
  • the invention provides for methods and pharmaceutical compositions to achieve an abuse deterrent formulation of abusable drugs.
  • the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release and abuse deterrence for abusable drugs.
  • the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release and abuse deterrence for abusable drugs, using substantially the same excipients. [00589] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve abuse deterrence, without the use of aversive agents. [00590] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to simultaneously achieve extended release and abuse deterrence, without the use of aversive agents.
  • the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release formulation and an abuse deterrence formulation, using substantially the same ADER agents.
  • the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents.
  • the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention providing abuse deterrence without the use of aversive agents.
  • the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention using substantially the same ADER agents to achieve the foregoing (i) to (v).
  • the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention using substantially the same ADER agents to achieve the foregoing (i) to (v); said invention providing abuse deterrence without the use of aversive agents.
  • the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and at least two ADER agents.
  • the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and at least two ADER agents; said invention providing abuse deterrence without the use of aversive agents.
  • the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and at least two ADER agents; said invention using substantially the same ADER agents to achieve the foregoing (i) to (v).
  • the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release formulation and an abuse deterrence formulation, using substantially the same
  • the pharmacologic effects of the pharmaceutical compositions of the present invention can be evaluated using methods well established in the art. The choice of method will depend, among other things, on: (i) the abusable drug and (ii) the therapeutic use to which the abusable drug is applied (i.e., the disease, disorder, or symptom(s) being treated with the abusable drug).
  • Some abusable drugs for example, cannabinoid agonists, stimulant drugs, or benzodiazepines have multiple therapeutic applications.
  • certain evaluations may be conducted in healthy subjects, recreational drug users or drug addicts. A wide variety of clinical states and study designs may be used to evaluate the therapeutic effects of intact and tampered dosage forms of the invention.
  • Pain intensity (VAS and categorical), pain relief (categorical) and whether pain is half-gone is recorded by the patient under the supervision of the investigator study coordinator at the various time points: Baseline (0 hour - pain intensity only), 15, 30 and 45 minutes, and at 1, 1.5, 2, 3, 4, 5, 6, 7, 8 and 12 hours after administration of study medication, and immediately prior to the first rescue dose. Sedation and nausea may be evaluated using VAS or categorical scales. Time to onset of perceptible and meaningful pain relief is evaluated using the two stopwatch method. Patients record their global evaluation of study medication at the completion of the 8-hour assessment or at the time of first rescue medication use.
  • Efficacy endpoints include Total Pain Relief (TOTPAR), Sum of Pain Intensity Difference (SPID) and Sum of Pain Relief Intensity Difference (SPRID) at various time points, Time to First Rescue, Time Specific Pain Intensity Difference (PID), Time Specific Pain Relief (PR), Peak Pain Intensity Difference (PPID) 5 Peak Pain Relief (PPR), Time to Confirmed Perceptible Pain Relief (stopwatch), Time to Meaningful Pain Relief (stopwatch), Patient Global Evaluation, Time to Change in Categorical PID > 1, Percent Change in Pain Intensity Score from Baseline, Mean Change in Pain Intensity Score From Baseline, Percent Change in Pain Relief Score from Baseline, Mean Change in Pain Relief Score From Baseline, Percent of Responders, Number of Patients Needed to Treat to Obtain One Patient with > 50% Response (NNT).
  • TOTPAR Total Pain Relief
  • SPID Sum of Pain Intensity Difference
  • SPRID Sum of Pain Relief Intensity Difference
  • both single and multiple (repeated) dose studies may be conducted. Patients are encouraged to wait at least 60 minutes before requesting remedication for pain. At the completion of the single-dose phase (S hours) or at first request for remedication (whichever is earlier), patients enter into a multiple-dose phase lasting approximately 72 hours. During the multiple dose phase patients receive study medication or placebo at a fixed dose interval (e.g., every 8, 12 or 24 hours).
  • Pain intensity Pain intensity (VAS and categorical), pain relief (categorical) and whether pain is half-gone is recorded by the patient under the supervision of the investigator study coordinator at representative time points, e.g., Baseline (pain intensity only), 15, 30 and 45 minutes and 1, 1.5, 2, 3, 4, 5, 6, 7 and 8 hours after administration of study medication and immediately prior to the first remedication. Sedation and nausea may be evaluated using VAS or categorical scales. Time to onset of perceptible and meaningful pain relief is evaluated using the double-stopwatch method. Patients complete a global evaluation of study medication at the completion of the 8-hour assessment or just prior to the first remedication.
  • patients Following completion of the single-dose phase (8 hours or just prior to first remedication, if ⁇ 8 hours), patients begin the multiple dose phase of the study. During the multiple dose phase, patients record their overall pain intensity since the previous scheduled dose, their current pain intensity and a patient global, immediately prior to each scheduled dose of study medication and at early termination.
  • SPID Total Pain Relief
  • SPRID Sum of Pain Relief Intensity Difference
  • PID Time Specific Pain Intensity Difference
  • PR Time Specific Pain Relief
  • PJD Peak Pain Intensity Difference
  • PPR Peak Pain Relief
  • Stopwatch Time to Meaningful Pain Relief
  • PID Percent Change in Categorical PID > 1, Percent Change in Pain Intensity Score from Baseline, Mean Change in Pain Intensity Score From Baseline, Percent Change in Pain Relief Score from Baseline, Mean Change in Pain Relief Score From Baseline, Percent of Responders, Number of Patients Needed to Treat to Obtain One Patient with > 50% Response (NNT).
  • Measures of efficacy in the multiple-dose phase include the time specific overall pain intensity, current pain intensity and patient global at the time of scheduled remedication, the average of overall pain intensity, current pain intensity and patient global over 0-24, 24-48 and 48-72 and number of doses of rescue analgesic over 0-24, 24-48 and 48-72 and 0-72 hours.
  • the analgesic efficacy of the invention may be demonstrated in single or repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. In repeated dose studies, typically, patients who meet the American College of Rheumatology criteria for knee and/or hip OA are washed off their analgesics for 2 to 7 days to allow for pain of moderate to severe intensity to return. Once a stable baseline pain score is established, patients are randomized to treatment, usually for a period of 1 to 12 weeks.
  • Pain, joint stiffness and physical function can be measured with a multidimensional instrument, such as the WOMAC, quality of life with the SF- 12 or SF-36 and adverse events with a non-directed questionnaire at baseline and at post-baseline return visits. Response to pain, stiffness, physical function, quality of life and adverse events are calculated as change from baseline and compared between treatments. Sedation and nausea may be evaluated using VAS or categorical scales.
  • the analgesic efficacy of the invention may be demonstrated in single or repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Patients with migraine headaches are typically evaluated in prospective, randomized, double-blind, parallel group, single-dose studies. Crossover studies are also possible. The study population consists of male and non-pregnant female subjects, 18 to 65 years of age with a primary headache diagnosis of either migraine attack without aura or migraine attack with aura, as diagnosed according to the International Classification of Headache Disorders-2 criteria.
  • the subject must typically have a history, on average, of at least one migraine attack per month, but an average of no more than 6 migraine attacks each month during the past year.
  • subjects are instructed to treat and evaluate the headache pain and symptoms associated with one eligible migraine attack, with or without aura, with at least moderate headache pain intensity.
  • Eligible subjects are randomly assigned to receive the drug to treat one migraine attack, with or without aura, with headache pain of at least moderate pain intensity as determined by them migraine questionnaire they are asked to take a single dose of study drug, according to their randomized treatment assignment. Headache pain intensity, nausea, photophobia, phonophobia, vomiting, and ability to function are assessed at baseline, 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, 8, 16 and 24 hours post-dose.
  • Primary efficacy variables typically consist of the percent of subjects who are without: (i) pain; (ii) nausea; (iii) photophobia and, (iv) phonophobia, each at 2 hours post-dosing.
  • Secondary efficacy variables typically consist of headache pain intensity and associated symptoms at each evaluation time point, incidence of vomiting, patient function, sum of pain intensity difference at each evaluation time (SPED), percent of subjects who experience headache recurrence up to 24 hours, and the median time to recurrence. Sedation may be evaluated using VAS or categorical scales.
  • Recurrence is defined as the reduction in pain from moderate or severe pain to none at 2 hours after taking study drug, followed by: (i) an increase to mild, moderate or severe pain within 24 hours after taking the study drug, or (ii) consuming a rescue medication within 24 hours after taking the study drug.
  • the analgesic efficacy of the invention may be demonstrated in repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Patients with a history of postherpetic neuralgia > 3 months and pain of at least moderate intensity are enrolled in the study. Patients with hypersensitivity to study medications, a history of drug or alcohol abuse and significant pain of alternate etiology are generally excluded. Patients meeting study eligibility criteria are "washed off their analgesics in some embodiments, generally for 2 to 7 days to allow for pain of moderate to severe intensity to return.
  • Pain intensity is assessed one to several times a day and in some cases only once weekly using VAS, categorical or numerical rating scales.
  • Various dimensions of neuropathic pain may be assessed, including steady pain (ongoing pain), brief pain (paroxysmal pain) and skin pain (allodynia). Pain may also be assessed at scheduled clinic study visits.
  • Pain may also be assessed using standardized pain scales such as the Neuropathic Pain Scale (Galer et al., Neurology 1997;48:332-8), the Neuropathic Pain Symptom Inventory (Bouhassira et al., Pain 2004;108:248-57), interference measures of the Brief Pain Inventory (Cleeland, CRC Press, 1991:293-305 and Ann Acad Med Singapore 1994;23: 129-38) or the McGiIl Pain Questionnaire Short- Form (Melzack, Pain 1987;30:191-7).
  • Patient global assessment may be measured using a number of available tools, for example Patient Global Impression of Change (Farrar et al., Pain 2001 ;94: 149-580).
  • Quality of life may similarly be assessed using number of available tools, for example the SF-36, SF-12 or SF-8.
  • Examples of randomized, placebo or active studies conducted in postherpetic neuralgia are known in the art (e.g., Watson and Babul, Neurology 1998;50:1837-41; Sabatowski et al., Pain. 2004;109:26-35; Rowbotham et al., JAMA. 1998;280: 1837-42).
  • Adverse events may be assessed using a non-directed questionnaire, a symptom checklist or specific queries on adverse signs and symptoms. Response to pain, function, quality of life and adverse events are calculated as change from baseline and compared between treatments. Sedation and nausea may be evaluated using VAS or categorical scales.
  • ADHD ' Attention-deficit/hyperactivity disorder
  • the efficacy and safety of the invention may be demonstrated in repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Patients are usually titrated to optimal doses. Adults or children with a history of ADHD meeting DSM-IV-TR criteria for ADHD are enrolled in the study. Patients with hypersensitivity to study medications, a history of drug or alcohol abuse and significant co-morbid pathology that could confound ADHD assessment are generally excluded.
  • Outcome measures for adult ADHD studies may include the Wender-Reimherr Adult Attention Deficit Disorder Scale (WRAADDS), the adult ADHD-Rating Scale (ADHD- RS), the Conners' Adult ADHD Rating Scale Short Version Self-Report (CAARS-S-S), and the Clinical Global Impressions-Improvement scale (CGI- I).
  • WRAADDS Wender-Reimherr Adult Attention Deficit Disorder Scale
  • ADHD-RS adult ADHD-Rating Scale
  • CAARS-S-S Conners' Adult ADHD Rating Scale Short Version Self-Report
  • CGI- I Clinical Global Impressions-Improvement scale
  • Outcome measures in children may include the teacher-completed Swanson, Nolan, and Pelham Rating Scale (Teacher SNAP), parent-completed SNAP (Parent SNAP) 5 Clinical Global Impressions Scale-Improvement (CGI- I) score, Math Test performance, the Teacher and Parent ADHD Rating Scale, the Conners' ADHD/DSM-IV Scales for teachers (CADS-T) and for parents (CADS-P).
  • Patients are randomized to treatment, usually for a period of 2 to 12 weeks.
  • Adverse events may be assessed using a non-directed questionnaire, a symptom checklist or specific queries on adverse signs and symptoms. Response may be calculated as change from baseline and compared between treatments.
  • the efficacy and safety of the invention may be demonstrated in repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Aged 21 to 69 years meeting DSM-IV-TR criteria for primary insomnia and reporting less than 6.5 hours of sleep per night, and/or a sleep latency of more than 30 minutes each night for at least 1 month before screening are enrolled in the study. Patients with hypersensitivity to study medications, a history of drug or alcohol abuse and significant co-morbid pathology that could confound sleep assessment are generally excluded.
  • Outcome measures for may include Efficacy and next-morning effects may be evaluated via polysomnography, Digit Symbol Substitution Test and self- report.
  • Study endpoints may include sleep latency, total sleep time, number of awakenings, wake time after sleep onset, quality of sleep and next-day ratings of ability to function, daytime alertness, and sense of physical well-being.
  • Patients are randomized to treatment, usually for a period of 2 to 6 weeks.
  • Adverse events may be assessed using a non-directed questionnaire, a symptom checklist or specific queries on adverse signs and symptoms. Response may be calculated as change from baseline and compared between treatments.
  • the popularity of immediate release formulations of abusable drugs among drug addicts and recreational drug users is in part due to the mood altering and reinforcing effects of the drug.
  • the popularity of extended release formulations of abusable drugs among drug addicts and recreational drug users is in part due to the pharmacologic properties of the drug therein (e.g., mood altering and reinforcing effects) and in part due to the large amount of drug per tablet or capsule (e.g., a 12 or 24 hour supply).
  • pharmacologic properties of the drug therein e.g., mood altering and reinforcing effects
  • the large amount of drug per tablet or capsule e.g., a 12 or 24 hour supply.
  • commercially available immediate release abusable drugs tablets and capsules are usually administered every 4 to 6 hours and they release their dose into the systemic circulation over one to two hours.
  • New, extended release formulations are designed to gradually release their much larger abusable drugs content over a 12 or 24-hour period.
  • Most recreational drug users and addicts have a unit of use which is one tablet or capsule.
  • the 12 or 24-hour supply of an abusable drugs typically contained in one extended release tablet or capsule, instead of in 4 to 6 tablets or capsules means that there is a greater risk that such formulations may be highly sought by drug addicts and recreational drug users alike, for non-medical use. Intentional or inadvertent tampering from extended release formulations will rapidly deliver a massive dose and produce profound pharmacologic effects.
  • parenteral e.g., intravenous injection, where the drug is crushed and extracted or melted and the contents of a dosage unit then injected
  • intranasal e.g., snorting, where the drug is inhaled as powdered dosage unit
  • episodic or repeated oral ingestion of crushed product where
  • One mode of abuse involves the extraction of the drug component from the dosage form by first mixing the tablet or capsule with a suitable solvent (e.g., water or alcohol), and then filtering and/or extracting the drug component from the mixture for intravenous injection.
  • a suitable solvent e.g., water or alcohol
  • Another mode of abuse of extended release drugs involves dissolving the drug in water, alcohol or another "recreational solvent" to hasten its release and to ingest the contents orally, in order to provide. high peak concentrations and maximum euphoriant effects.
  • Method Place a whole dosage unit in 18 mL of 0.1N HCl in a 60 mL amber bottle and shake at 240 rpm on an orbital shaker for 30 min. After 30 min add 12 mL of ethanol (95-96%) to each bottle. Swirl by hand and remove a 1 mL sample from each bottle (To). Place the solutions back in the orbital shaker for further shaking at 240 rpm. Take 1 mL samples after 10, 20, 30, 40, 60 and 180 min of further shaking for each bottle. Analyze and graph the results on a linear scale of cumulative release (%) vs. time (min).
  • Method Freeze the dosage unit in a domestic freezer for 24 hr, then grind with a mortar and pestle for five minutes. Sieve through a suitable sieve (ca ⁇ OOmicron) and, by weighing, measure the percentage passing the sieve.
  • Method Crush with a mortar and pestle and heat to boiling in 5 mL of vinegar. The resulting suspension is filtered through a 0.45 micron filter into a flask and diluted to 50 mL with water. Quantify drug concentration by HPLC.
  • Method Heat the squashed contents of a dosage unit on a hot plate until melted. Determine the temperature of melting and test whether the mix becomes sufficiently fluid to be drawn up into a syringe via a 1.2 mm needle then expelled.
  • Non-limiting examples for preparing the dosage form are set forth below.
  • Suitable abusable drugs as defined in this invention may also be prepared by modification of the examples herein and by use of material other than those specifically disclosed herein, including those which may hereafter become known to the art to be capable of performing the necessary functions.
  • Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered and obvious to those skilled in the art are within the spirit and scope of the invention.
  • More than abusable drug may be included in the dosage form either in an immediate release or extended release form
  • the ingredients used for the preparation of the dosage form may be modified depending on the selection, dose and desired duration of effect of the abusable drug and any included aversive agent, pharmacologic antagonist or other non-abusable therapeutic agent.
  • a change in the dose or amount of abusable drug does not require a change in amount of other ingredients.
  • a proportional change in the amount of other ingredients is required to maintain the desired properties.
  • a change in the dose or amount of abusable drug necessitates a change in the nature and/or amount of ingredients to provide the required characteristics of the abusable drug (e.g., immediate release, sustained release, duration of effect, rate and extent of absorption, therapeutic concentrations and effect, abuse deterrence properties, protection against alcohol dose dumping; and/or protection against significant changes in bioavailability due to fed or fasted states etc.)
  • aversive agents may be included sleeted from the group comprising (i) laxatives; (ii) cutaneous vasodilators; (iii) headache producing agents; (iv) emetics, emetogenic and nausea producing compounds; (iv) bittering agents (v) mucosal, naso-mucosal, oro-mucosal, respiratory, tissue and gastrointestinal irritants; (vi) tissue staining, non-tissue staining and beverage staining dyes, lakes and colorants; (vii) fecal and urine discolorants; (viii) malodorous agents; (ix) opioid antagonists; and (x) and (x) benzodiazepine antagonists (e.g., flumazenil), and mixtures thereof.
  • laxatives comprising (i) laxatives; (ii) cutaneous vasodilators; (iii) headache producing agents; (iv) emetics, emetogenic and
  • Manufacturing methods described herein are utilized for the preparation of the abusable drugs as shown in the examples below. Variations to the methods may be employed, in some embodiments, depending on the specific chemical, physicochemical, pharmaceutical and pharmacologic properties of the abusable drug, excipients and their interaction and other factors.
  • any abusable drug of the invention may be prepared to provide (i) abuse deterrence; or (ii) extended release; or (iii) resistance against alcohol dose dumping; or (iv) resistance against alcohol dose dumping when formulated as extended release; or (v) protection against significant changes in bioavailability due to fed or fasted states; or (vi) more than one of the foregoing [(i) to (v)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention, in some embodiments, providing further abuse deterrence through the use of aversive agents; said invention, in some embodiments, using substantially the same ADER agents to achieve the foregoing (i) to (v); said invention, in some embodiments, providing more than one of the foregoing [(i) to (v)] properties; said invention, in some embodiments, comprising one or more abusable drugs and at least two ADER agents; said invention, in some embodiments, comprising one or more abusable drugs and at
  • mp 45 to 100
  • Example 16 to 150 may be prepared as using the formula of Examples

Abstract

The present invention is in the field of oral, abuse resistant pharmaceutical compositions of abusable drugs, extended release pharmaceutical compositions of abusable drugs and extended release abuse resistant pharmaceutical compositions of abusable drugs and the use thereof. The present invention is also directed to extended release pharmaceutical compositions and the use thereof for preventing or minimizing the risk of abuse and/or toxicity from either intentional or unintentional tampering. The present invention is further directed at a method of preventing or minimizing the risk of abuse and/or toxicity from either intentional or unintentional tampering.

Description

MULTIMODAL ABUSE RESISTANT AND EXTENDED RELEASE
FORMULATIONS
FIELD OF THE INVENTION
[0001] The present invention is in the field of multimodal oral, abuse resistant pharmaceutical compositions of abusable drugs, extended release pharmaceutical compositions of abusable drugs, extended release abuse resistant pharmaceutical compositions of abusable drugs and the use thereof.
BACKGROUND OF THE INVENTION
[0002] Although the abuse of unregulated psychoactive or mood altering naturally occurring and synthetic chemical substances and "street drugs" has been documented over many centuries, it is largely in the last century that we have been confronted by the abuse of pharmaceutical grade psychoactive or mood altering substances in the form of pharmaceutical dosage forms intended for therapeutic uses. The abuse of pharmaceutical products is either due to iatrogenic addiction or willful abuse of the products outside their intended use or method of use by drug abusers and recreational drug users. Most of the abuse of pharmaceutical products, regardless of etiology, is confined to substances that have pleasurable, mood altering, rewarding and/or reinforcing effects.
[0003] A wide variety of pharmaceutical products can be subjected to abuse including psychostimulants, amphetamine and amphetamine analogs, amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, barbiturates and cannabinoid agonists. Some of the abusable substances within these classes of drugs are currently available for commercial sale to prevent or treat medical conditions, while others are in development.
[0004] On its website, the National Institutes of Drug Abuse (NIDA) notes
"Prescription drugs that are abused or used for nonmedical reasons can alter brain activity and lead to dependence. Commonly abused classes of prescription drugs include opioids (often prescribed to treat pain), central nervous system depressants (often prescribed to treat anxiety and sleep disorders), and stimulants (prescribed to treat narcolepsy, ADHD, and obesity). Commonly used opioids include oxycodone (OxyContin™), propoxyphene (Darvon™), hydrocodone (Vicodin), hydromorphone (Dilaudid™), meperidine (Demerol™), and diphenoxylate (Lomotil™). Common central nervous system depressants include barbiturates such as pentobarbital sodium (Nembutal™), and benzodiazepines such as diazepam (Valium™) and alprazolam (Xanax™). Stimulants include dextroamphetamine (Dexedrine™) and methylphenidate (Ritalin™). Long- term use of opioids or central nervous system depressants can lead to physical dependence and addiction. Taken in high doses, stimulants can lead to compulsive use, paranoia, dangerously high body temperatures, and irregular heartbeat." (www.nida.nih.gov, accessed September 7, 2006)
[0005] The major active chemical in marijuana is delta-9- tetrahydrocannabinol (THC), which causes the mind-altering effects of marijuana intoxication. The amount of THC (which is also the psychoactive ingredient in hashish) determines the potency and, therefore, the effects of marijuana. Between 1980 and 1997, the THC content of marijuana available in the United States rose dramatically.
[0006] Marijuana is the most commonly used illicit drug in the United States.
More than 94 million Americans (40 percent) age 12 and older have tried marijuana at least once, according to the 2003 National Survey on Drug Use and Health (NSDUH). Marijuana use is widespread among adolescents and young adults. It is reported that in 2004, 16 percent of 8th-graders volunteered that they had tried marijuana, and 6 percent were current users (defined as having used the drug in the 30 days preceding the survey). Among 10th- graders, 35 percent had tried marijuana sometime in their lives, and 16 percent were current users. As would be expected, rates of use among 12th-graders were higher still. Forty-six percent had tried marijuana at some time, and 20 percent were current users. [0007] The Drug Abuse Warning Network (DAWN), a system for monitoring the health impact of drugs, estimated that, in 2002, marijuana was a contributing factor in over 119,000 emergency department (ED) visits in the United States, with about 15 percent of the patients between the ages of 12 and 17, and almost two-thirds male.
[0008] In 2002, the National Institute of Justice's Arrestee Drug Abuse
Monitoring (ADAM) Program, which collects data on the number of adult arrestees testing positive for various drugs, found that, on average, 41 percent of adult male arrestees and 27 percent of adult female arrestees tested positive for marijuana. On average, 57 percent of juvenile male and 32 percent of juvenile female arrestees tested positive for marijuana.
[0009] Cannabis use is world-wide public health issue. According to the
United Nations Office for Drug Control and Crime Prevention (UNODCCP), Marijuana is the most widely used illicit drug in the world. It has been estimated that one in 11 cannabis users will become dependent (Anthony et al., Clin Psychopharmacol, 1994); rates of cannabis dependence in several countries (e.g., Australia, USA, South Africa) have increased substantially over the past decade (Bhana et al., S Afr Med J, 2002; SAMHSA, 2003), as well as the number of individuals seeking treatment (Stephens et al., Clin Psychol, 1993; Treatment Episode Data Set, 2002).
[0010] The principal psychoactive constituent of marijuana, THC, was not definitively identified until 1964 (Gaoni and Mechoulam, J Am Chem Soc, 1964). Unfortunately, marijuana is not a good source of THC due to the difficulties in isolation and purification. The development of a practical synthetic pathway (Razdan et al., Experientia, 1972) was a major boost to the subsequent pharmacological characterization of the effects of marijuana and synthetic cannabinoids. Advances in chemistry, behavioral pharmacology, molecular pharmacology and neurobiology have facilitated the identification and characterization of an endogenous cannabinoid system.
[0011] A number of pathologic states affect the human cannabinoid system, including Alzheimer's disease, schizophrenia, depression, alcoholism, Parkinson's disease, stroke, premature labor, endotoxic shock, hepatic cirrhosis, atherosclerosis, cancer, bone implantation, glaucoma, emesis, and pruritus of various etiology (Pertwee, AAPS Journal, 2005; Mackie Annu Rev Pharmacol Tox, 2006).
[0012] There are also signs of upregulation or downregulation of the endocannabinoid system in a variety of animal in vivo models, including multiple sclerosis, amyotrophic lateral sclerosis, encephalitis, Alzheimer's disease, Parkinson's disease, Huntington's disease, obesity, feeding, fasting, stress, memory, aging, hypertension, cirrhosis, septic shock, cardiogenic shock, cerebral ischemia, myocardial infarction, neurotoxicity, febrile seizures and various intestinal disorders (Pertwee, AAPS Journal, 2005).
[0013] There exists considerable concern on the part of public health policy experts, addiction medicine specialists, pharmaceutical companies and government health regulators about the potential risk of drug abuse and drug diversion with the commercialization and widespread use of new cannabinoid agonists currently in development. Pharmaceutical companies have responded to the risk of abuse from cannabinoid agonists in different ways.
[0014] Some pharmaceutical companies with significant expertise in cannabinoid chemistry and discovery have elected to focus on therapeutic potential of cannabinoid antagonists to the exclusion of cannabinoid agonists.
[0015] Other pharmaceutical companies have decided to focus on CB2 cannabinoid agonists since it is assumed that CB2 receptors are found mainly outside of the brain, in immune cells and that CBj receptors are found throughout the body, but primarily in the brain.
[0016] An important drawback with the use of cannabinoid agonist is the risk of drug addiction, drug diversion and drug abuse. For instance, unsuspecting clinicians may prescribe a cannabinoid agonist to patients who have an underlying, undisclosed addiction disorder. Alternatively, unsuspecting clinicians may prescribe a cannabinoid agonist to patients with a medical condition amenable to treatment or prevention with a cannabinoid agonist who in turn divert a portion of their prescribed dose to other individuals for nonmedical use. [0017] There have also been documented cases of inappropriate prescribing or dispensing of psychoactive drugs by physicians and pharmacists, with its eventual diversion into the non-medical marketplace. Additionally, experience with other classes of psychoactive, abusable drugs suggest that non-medical supplies of pharmaceutical grade cannabinoid agonist will be available through prescription forgeries and break-ins into pharmacies.
[0018] Scheduling of cannabinoid agonist would have the unintentional consequence of causing physicians, fearful of being accused of permitting "overuse", to prescribe suboptimal doses of the drugs to patients in need of them, and to prescribe less effective drugs to patients that are not similarly scheduled. We have coined this phenomenon as "cannabinophobia" and "cannabiphobia".
[0019] Pharmaceutical dosage forms containing cannabinoid agonists will likely be used for non-medical purposes in a variety of settings: i) by patients with a medical condition amenable to treatment or prevention with a cannabinoid agonist who have developed an addiction disorder following initiation of the cannabinoid agonist; ii) by patients with an addiction disorder seeking cannabinoid agonists for their euphoriant properties and iii) by recreational drug users who may use cannabinoid agonists from time to time ("chippers") for pleasure seeking effects, analogous to the intermittent use of marijuana by many users.
[0020] Cannabinoids hold substantial promise for the prevention and treatment of a wide variety of medical conditions, including multiple sclerosis, amyotrophic lateral sclerosis, encephalitis, Alzheimer's disease, Parkinson's disease, Huntington's disease, obesity, feeding, fasting, stress, schizophrenia, depression, alcoholism, stroke, premature labor, endotoxic shock, hepatic cirrhosis, atherosclerosis, pruritus of various etiology and cancer.
[0021] Therefore cannabinoid agonists are going to be widely used for a variety of pathologic states.
[0022] In view of its anticipated widespread use, cannabinoid agonists also have the potential to create a major epidemic of drug abuse involving an entirely new pharmacologic class of agents. [0023] Epidemiologic data suggest that pharmaceutical cannabinoid agonists are also likely to be co-abused with alcohol.
[0024] . There is a need, therefore, for novel methods of deterring or preventing cannabinoid agonist abuse.
[0025] There is a need, therefore, for novel methods and pharmaceutical compositions of cannabinoid agonists that have abuse deterrence properties to deter their abuse and diversion.
[0026] There is a need, therefore, for novel methods and pharmaceutical compositions of cannabinoid agonists in extended release form that have abuse deterrence properties.
[0027] With few exceptions, commercially available extended release formulations of pharmaceuticals in the United States are in the form of oral solid dosage forms (exceptions being some liquid formulations involving ion exchange resins). A number of cannabinoid agonists are liquid, semisolid or oily substances which are difficult to formulate into extended release formulations using conventional technology. The present invention is well suited to cannabinoid by providing a liquid fill, solid dispersion systems that provide abuse deterrence, extended release and protection against alcohol dose dumping.
[0028] Another abused pharmaceutical product is methylphenidate, a drug prescribed primarily in children and adolescents for the treatment of attention- deficit hyperactivity disorder (ADHD). Methylphenidate is a CNS stimulant with effects similar to, but more potent than, caffeine and less potent than . amphetamines. It has a paradoxical effect of calming and "focusing" efforts in individuals with ADHD, particularly children. Methylphenidate is an important therapeutic option for adults as well as children with ADHD. Due to its stimulant properties, however, in recent years there have been reports of methylphenidate abuse by people for whom it is not prescribed. It is abused for its stimulant effects: appetite suppression, wakefulness, increased focus/attentiveness, and euphoria. Addiction to methylphenidate seems to occur when it induces rapid increases of dopamine in the brain. In contrast, the therapeutic effect is achieved by slow and steady increases of dopamine, which are similar to the natural production by the brain. When abused, the tablets are either taken orally or crushed and snorted. Some abusers dissolve the tablets in water and inject the mixture; complications can arise from this because insoluble fillers in the tablets can block small blood vessels. (Source: Adapted from www.nida.nih.gov, accessed September 7, 2006)
[0029] Methylphenidate has been shown to serve as a positive reinforcer in monkeys (Johanson and Schuster, 1975, J Pharmacol Exp Ther; Wilson et al., 1971, Psycopharmacol), and has established potential for abuse (Wang et al., 1997, Eur Addict Res). In one study, methylphenidate consistently maintained a cocaine response in an animal model of cocaine addiction predictive of abuse potential in humans (Wee and Wolverton, Drug and Alcohol Dependence, 2004).
[0030] Methylphenidate is frequently the subject of theft from pharmacies and drug wholesalers. Organized drug-trafficking groups utilize various schemes to obtain methylphenidate for sale on the illicit market. There is also considerable diversion of methylphenidate. In one study, children and adolescents who had been prescribed methylphenidate were surveyed. The study found that approximately 20% of patients had been approached to sell, provide at no cost or trade their medicine at least once in the previous five 5 years (Musser et al., 1998).
[0031] Other epidemiological surveys indicate that methylphenidate abuse may be increasing. For example, the Indiana Prevention Resource Center reported that 7.5% of Indiana high school seniors had used methylphenidate recreationally, a rate comparable to cocaine use in this population. Babcock and Byrne (2000) reported that more than 16% of students at a college had experimented with methylphenidate recreationally. Additionally, approximately 13% of students had taken methylphenidate intranasally. These data are similar to the prevalence of recreational use of cocaine and amphetamine on the same campus. In a recent survey, the American Association of Poison Control Centers reported that over 11% of reported methylphenidate exposures during a 7-year period involved intentional abuse (Klein-Schwartz and McGrath, 2003). Of these cases, over 20% involved intranasal use of methylphenidate. Several case studies have also reported abuse of methylphenidate, especially via the intranasal route (Coetzee et al., 2002; Jaffe, 1991, 2002; Garland, 1998; Massello and Carpenter, 1999). In general, the reports indicate that adolescents are pulverizing a few methylphenidate tablets and administering the powder intranasally. Stoops et al. have reported that intranasal methylphenidate produces linear dose- response prototypical stimulant-like subjective effects (e.g. increases in ratings of Good Effects and High) and concluded that across a range of doses, intranasal methylphenidate produces behavioral effects that are characteristic of abused stimulants (Stoops et al., Drug and Alcohol Dependence, 2003).
[0032] Similar patents of abuse have been observed for amphetamine, amphetamine analogs, amphetamine like drugs, other psychostimulants and some anorectics.
[0033] With the increasing recognition of ADHD in children and adults and the expanding application of stimulant drugs for new indications, these agents are going to be used even more widely for a variety of pathologic states.
[0034] With the increasing recognition of the utility of putative agents for the treatment of obesity, excessive daytime sleepiness and other disorders, amphetamine, amphetamine analogs, amphetamine like drugs, psychostimulants and anorectics are going to be used even more widely for a variety of pathologic states.
[0035] There is a need, therefore, for novel methods and pharmaceutical compositions of stimulants in extended release form that provide a prolonged duration of action, with a reduced frequency of peak to trough fluctuations and improved compliance.
[0036] There is also a need, therefore, for novel methods and compositions for deterring or preventing stimulant abuse.
[0037] There is also a need for novel methods and compositions for deterring or preventing stimulant abuse when said agonist is formulated as an extended release formulation. [0038] There is also a need for novel methods and compositions for deterring or preventing stimulant abuse when said agonist is formulated as an extended release formulation.
[0039] Another class of drugs that have significant potential for abuse, misuse and diversion are the benzodiazepines. In a study with squirrel monkeys intravenous midazolam maintained high and consistent rates of responding (Munzar et al., 2001). In three other studies, benzodiazepines served as effective reinforcers for rhesus monkeys and provide further evidence that benzodiazepines share significant characteristics with other drug reinforcers. (Gomez et al, 2002; Gomez et al., 2002; Stewart et al., 1994).
[0040] Epidemiological studies and surveillance reports indicate that the nonmedical use of prescription benzodiazepines agonists or benzodiazepines is increasing significantly among young adults and college students in the U.S. (e.g., Johnston et al., 2003; Mohler-Kuo et al., 2003; SAMHSA, 2002 and 2003). Benzodiazepines are widely used anxiolytic medications due to their established efficacy and safety for the treatment sleep and anxiety disorders. There has been a recent increase in the U.S. prescription rates for benzodiazepines in general (Pincus et al., 1998) and for benzodiazepine anxiolytics among youths 20 years and adolescents (Zito et al., 2003). Although benzodiazepines are safe and effective when used as intended, their increased use has raised public health concerns because of they also carry a high propensity for abuse. Additionally, the nonmedical use of benzodiazepines among adolescents and young adults has been on the increase (Gledhill-Hoyt et al., 2000; Johnston et al., 2003; Mohler-Kuo et al., 2003; SAMHSA, 2002). The National Survey on Drug Use and Health (NSDUH) reported in 2001 data which indicated that young adults 18 to 25 years of age had the highest prevalence of nonmedical use of prescription drugs (SAMHSA, 2002). The lifetime nonmedical use of several benzodiazepines increased significantly (e.g., Valium from 5.4% to 6.3%) between 2000 and 2001 among those U.S. young adults 18-25 years of age (SAMHSA, 2002).
[0041] The Drug Abuse Warning Network (DAWN) has also reported a recent increase in the abuse of prescription benzodiazepines. According to DAWN, emergency department mentions of benzodiazepines significantly increased (38%) from 1995 to 2002. In particular, mentions of alprazolam (Xanax™) increased 62% over the same time frame and mentions of clonazepam (Klonopin™) increased 33% (SAMHSA, 2003).
[0042] The Harvard School of Public Health College Alcohol Study (CAS) has similarly reported that the nonmedical use of benzodiazepines more than doubled between 1993 and 2001 (1.8% and 4.5%, respectively) in the past decade among U.S. college students (Mohler-Kuo et al., 2003).
[0043] There is a need, therefore, for novel methods of deterring or preventing benzodiazepines agonist abuse.
[0044] There is a, need, therefore, for novel methods and pharmaceutical compositions of benzodiazepines in extended release form that provide a prolonged duration of action, with a reduced frequency of peak to trough fluctuations and improved compliance.
[0045] There is a need, therefore, for novel methods and pharmaceutical compositions of benzodiazepines agonists that have abuse deterrence properties to deter their abuse and diversion.
[0046] There is a need, therefore, for novel methods and pharmaceutical compositions of benzodiazepines agonists in extended release form that have abuse deterrence properties.
[0047] Many important therapeutic applications of abusable drugs (e.g., cannabinoid agonists, benzodiazepine agonists, and stimulant drugs) are for the treatment of chronic conditions where continuous suppression of signs and symptoms is important. In certain conditions, this may be achieved by the use of extended release dosage forms that provide sustained therapeutic plasma concentrations.
[0048] There is a need, therefore, for novel methods and pharmaceutical compositions of extended release formulations of cannabinoid agonists to provide continuous relief of signs and symptoms amenable to treatment with cannabinoid agonists, without having to take frequent doses of short acting cannabinoid agonists. [0049] There is a need, therefore, for novel methods and pharmaceutical compositions of cannabinoid agonists in extended release form that provide a prolonged duration of action, with a reduced frequency of peak to trough fluctuations and improved compliance.
[0050] For many abusable drugs with short half-lives or short duration of action, conventional (so called "immediate-release", "rapid release" or "short acting") drugs require frequent dosing. In contrast, extended release oral dosage forms are designed to maintain effective plasma levels throughout an 8, 12 or 24-hour dosing interval. For many abusable drugs with short half- lives or short duration of action, extended release formulations have now become the standard of care. Use of extended release dosage forms can result in fewer interruptions in sleep, reduced dependence on caregivers, improved compliance, enhanced quality of life outcomes, and increased control over the management of their disease. In the case of certain conditions involving children (e.g., with ADHD), the very elderly or cognitively impaired, extended release formulations can be important in reducing the need for supervised dosing in school or at home. In addition, such formulations can provide more constant plasma concentrations and clinical effects, less frequent peak to trough fluctuations and fewer side effects, compared with short acting drugs.
[0051] An important drawback with the use of abusable pharmaceutical dosage forms is the risk of drug addiction, drug diversion and drug abuse. Although the use of botanicals and drugs for non-medical or non-therapeutic purposes has existed throughout recorded human history, their abuse has increased significantly in the past three decades (Drug Abuse Warning Network, http://dawninfo.samhsa.gov/; Drug Enforcement Administration, http://www.deadiversion.usdoj.gov/; National Survey on Drug Use & Health, http://www.oas.samhsa.gov/nhsda.htm; American Association of Poison Control Centers Toxic Exposure Surveillance System, http://www.aapcc.org/annual.httn).
[0052] Our increased understanding of the clinical pharmacology and data from well controlled clinical trials have resulted in more widespread use of "dual use" drugs (drugs with legitimate therapeutic potential but which also have abuse potential). This in turn has led to concerns about the increased non-medical use through both licit and illicit channels. For instance, unsuspecting clinicians may prescribe such abusable or dual use drugs for a legitimate medical use to individuals with an addiction disorder or individuals who divert a portion of their prescribed dose to other individuals. There have also been documented cases of inappropriate prescribing or dispensing of abusable drugs by physicians and pharmacists, with its eventual diversion into the non-medical marketplace. Additionally, non-medical supplies of pharmaceutical grade abusable drugs are often obtained through prescription forgeries and break-ins into pharmacies.
[0053] Pharmaceutical dosage forms containing abusable drugs may be ingested whole, crushed and ingested, crushed or vaporized and snorted or injected intravenously after attempted extraction of the active pharmaceutical ingredient.
[0054] The introduction of extended release forms of abusable drugs (e.g.,
ADHD, insomnia) has revolutionized the management of many chronic conditions. However, the widespread use of extended release dosage form of abusable drugs has also associated with its diversion into the non-medical supply for use both by addicts and recreational drug users.
[0055] The popularity of extended release oxycodone among addicts and recreational drug users was due to a large amount of drug per tablet (e.g., a 12 or 24 hour supply). Commercially available immediate release tablets and capsules usually release their dose into the systemic circulation over one to two hours. New, extended release formulations are designed to gradually release their much larger opioid content, usually over a 12 or 24-hour period. Most recreational drug users and addicts have a unit of use which is one tablet or capsule. For example, the 12 or 24-hour supply of an abusable drug contained in one tablet or capsule, instead of 4 to 6 tablets or capsules means that there is a greater risk that such formulations may be highly sought by drug addicts and recreational drug users alike, for non-medical use. Intentional or inadvertent tampering from extended release formulations will rapidly deliver a massive dose and produce profound a variety of serious and life threatening side effects, including, depending on the pharmacology of the abusahle drug, respiratory depression and stimulation, cardiac arrhythmias, sedation and hypersomnia, seizures, hypertension and hypertensive crises, cognitive and perceptual impairment, cardiovascular collapse, coma and death.
[0056] Addicts and recreational drug may use extended release abusable drugs by the parenteral, intranasal or oral route. Dosage form abuse can involve physical, mechanical, thermal or chemical tampering of the dosage form (e.g., crushing, melting, solvent extraction and filtration)
[0057] Scheduling of abusable drugs has also had the unintentional side-effect of causing physicians, fearful of being accused of permitting "drug overuse", to prescribe suboptimal doses of abusable drugs to patients in need of them, and to prescribe less effective drugs to patients that are not similarly scheduled.
[0058] Abuse deterrent formulations of abusable drugs that employ a sequestered antagonist can cause serious harm to patients by precipitating, in some cases, an abstinence syndrome through the liberation of the antagonist. There is therefore a need for a "passive" abuse deterrent system to protect both medical and non-medical users of abusable drugs from intentional or unintentional toxicity, without unnecessary harm to either group from the abuse deterrent technology.
[0059] Similarly, abuse deterrent pharmaceutical compositions containing aversive substances can, under certain conditions cause serious harm to subjects if injected intravenously and the long terms safety of small amounts of such aversive substances which would be inevitably released in the gastrointestinal tract is unknown.
[0060] There is also need, therefore, for novel methods of preventing abuse which do not require the incorporation of aversive and potentially unsafe agents into the formulation.
[0061] In 2005, a serious new clinical problem arose with the therapeutic use of extended release drugs, particularly extended release formulations in opioids in capsule dosage forms, when co-ingested with alcohol. In this setting, the opioid analgesic was being used for legitimate medical purposes (e.g., to treat pain) and was being ingested as an untampered or intact formulation. Although subjects with chronic pain are discouraged from using opioids with alcohol, the co-ingestion of opioids with alcohol, especially in the setting of intractable pain is widespread. The problem was discovered with a once-a-day extended release formulation of the opioid hydromorphone HCL (Palladone® capsules). Palladone® capsules were introduced in the United States and Canada in 2004. In 2005, Palladone® capsules were withdrawn from the market in both countries due to dose-dumping when co-ingested with alcohol. In a 24-subject study, patients consuming 240 mL of 40% ethanol had a 6-fold mean increase in peak plasma hydromorphone concentration compared with co-ingestion of Palladone® capsules with water. One subject experienced a 16-fold increase when the drug was ingested with 40% alcohol compared with water. Patients consuming 240 mL of 20% ethanol had a 2-fold mean increase in peak plasma hydromorphone concentration. One subject in this group experienced a 6-fold increase when the drug was ingested with 20% alcohol compared with water. In some subjects, 8 ounces of 4% alcohol (equivalent to 2/3 of a typical serving of beer) resulted in almost twice the peak plasma hydromorphone concentration than when the drug was ingested with water. In requesting the withdrawal of Palladone® capsules, FDA noted that the manufacturer of "Palladone® provided FDA data that showed that drinking alcohol while taking Palladone® capsules may cause rapid release of hydromorphone, leading to high drug levels in the body, with potentially fatal effects. High drug levels of hydromorphone may depress or stop breathing, cause coma, and even cause death. The Agency has concluded that the overall risk versus benefit profile of Palladone® is unfavorable due to a potentially fatal interaction with alcohol. Pharmacokinetic data indicate that the co-ingestion of Palladone® and alcohol results in dangerous increases in the peak plasma concentrations of hydromorphone. These elevated levels may be lethal, even in opioid tolerant patients." (Sloan and Babul, Expert Opinion on Drug Delivery 2006;3:489-97; http://www.fda.gov/cder/drug/infopage/palladone /default.htm) [0062] FDA has since noted that a number of other capsule formulations of extended release drugs may be similarly vulnerable to dose dumping when co- ingested with alcohol. In vitro studies performed by the FDA have demonstrated that when Avinza® (once-daily extended release morphine) 30 mg was mixed with 900 mL of buffer solutions containing ethanol, the dose of morphine that was released was alcohol concentration-dependent, leading to a more rapid release of morphine. While the relevance of in vitro lab tests regarding Avinza® to the clinical setting remains to be determined, this acceleration of release may correlate with in vivo rapid release of the total morphine dose, which could result in the absorption of a potentially fatal dose of morphine. (http://www.fda.gov/medwatch/SAFETY/2005/ AVINZA_DHCP_Letter_Oct 2005.pdf; Sloan and Babul, Expert Opinion on Drug Delivery 2006;3:489-97)
[0063] There is therefore also need, therefore, for novel methods of preventing excessive peak concentrations (dose dumping) of abusable drugs when they are co-ingested for medical purposes at prescribed doses with alcohol.
[0064] Extended release formulations have become highly preferable and in some cases, the standard of care for the management of a wide variety of conditions, particularly chronic conditions. Additionally, extended release formulations can make otherwise non-viable pharmaceutical agents (e.g., due to an exceedingly short duration of effect) into viable formulations with clinical and commercial potential.
[0065] Nonlimiting examples include abusable drugs for the management of:
(i) ADHD, which can provide continuous suppression of hyperactivity and inattention with morning parental or self-administration, without the need for supplemental (e.g., noon-time) dosing may a potentially non-compliant child or adolescent or the need for administration by a teacher or school nurse; and (ii) sleep, where sustained delivery may provide optimal control of insomnia with reduced sleep interruptions and improved polysomnography outcomes.
[0066] Toxicity from abusable drugs can result from unintentional or intentional tampering of abusable drugs. It is not uncommon for patients who have difficulty swallowing, to crush the contents of tablets or open a capsule, and swallow the contents with liquids or on soft food. In the case of most immediate release dosage forms of abusable drugs, this generally produces no significant harm, with marginally higher peak concentrations (Cmax) and time to peak concentrations (tmax). However, in the case of extended release dosage forms of abusable drugs, crushing the oral solid dosage form destroys the controlled-release mechanism and results in a rapid surge of drug into the bloodstream, with the entire 8, 12 or 24-hour drug supply released immediately with toxic effects, or pleasurable effects in the case of a drug abuser. For this reason, all extended release dosage forms available for sale in the United States carry a warning to the prescriber and patient not to crush or tamper with the oral solid dosage form.
[0067] There is a need, therefore, for novel methods and pharmaceutical compositions of extended release formulations of abusable drugs to provide continuous relief of signs and symptoms amenable to treatment with the abusable drug, without having to take frequent doses of abusable drugs. There is a need, therefore, for novel methods and pharmaceutical compositions of extended release formulations of abusable drugs that result in fewer interruptions in sleep, reduced dependence on caregivers, improved compliance, enhanced quality of life outcomes, and increased control over the management of their medical condition. There is a need, therefore, for novel methods and pharmaceutical compositions of extended release formulations of abusable drugs that result in more constant plasma concentrations and clinical effects, less frequent peak to trough fluctuations and fewer side effects, compared with short acting or immediate release versions of abusable drugs. There is a need for novel methods and pharmaceutical compositions of extended release formulations of abusable drugs that achieve the aforementioned benefits without increasing the risk of toxicity, drug diversion and drug abuse. There is a need for novel methods and pharmaceutical compositions of extended release formulations of abusable drugs that achieve the aforementioned benefits and also provide abuse deterrence.
[0068] To date, no extended release formulations of abusable drugs with abuse deterrent technology have been submitted for Marketing Application (New Drug Application) or been commercialized anywhere in the United States. Indeed if prior drug development history is any guide, most such strategies are unlikely to be developed or commercialized and the optimal formulation(s) will likely be apparent only through postmarketing surveillance of several formulations with competing technologies. In addition, regional differences in patterns of abuse mean that different abuse deterrence strategies may be useful in different part of the world. Finally, experience with substance abuses indicates that those who are habitual abusers, particularly those who inject drugs intravenously, have a remarkable ability to defeat abuse deterrence strategies through physical and chemical manipulation of drugs of abuse. Such addicts are frequently only one step behind strategies to deter abuse. With the ready access to information from their well knit network and more recently, from websites on how to optimally extract the active agent from pharmaceutical dosage forms and maximize euphoriant effects, the development of abuse deterrent formulations has become a major pharmaceutical, clinical, regulatory and law enforcement challenge. [0069] In view of this, it is not surprising that the Food and Drug
Administration's Division of Anesthetic, Analgesic and Rheumatology Drug Products and the U.S. Drug Enforcement Administration have encouraged companies to develop wide ranging abuse deterrent strategies for abusable drugs, particularly extended release abusable drugs and as "inducement", offered that such products may include in their prescribing information data about their products abuse deterrent properties (FDA Perspectives on Opioid Risk Management. Opioid Risk Management Meeting, Tufts Healthcare Institute, Boston, March 29, 2005; DEA Perspectives on Opioid Risk Management. Opioid Risk Management Meeting, Tufts Healthcare Institute, Boston, March 29, 2005).
[0070] Various attempts have been made and are described in prior art to develop abuse-deterrent dosage forms. Clearly there is a need for a delivery system for commonly used oral dosage formulations of drugs, for patients seeking drug, therapy and which deters abuse and minimizes or reduces the potential for psychological dependence. In particular, there is a need for formulations that simultaneously provide robust abuse deterrence properties and an extended release pharmacokinetic profile suitable for every 8, 12 or 24 hour oral administration. There is also a need for extended release formulations that are stable (i.e., do not dose dump) when used at therapeutic doses for medical purposes in conjunction with alcohol. An ideal formulation will provide a extended release pharmacokinetic profile suitable for every 8, 12 or 24 hour release and will be resist or frustrate attempts at crushing at room temperature and upon freezing, melting to allow for filtration and/or aspiration into a syringe and extraction with recreational solvents, all without doing harm to pain patients or patients with a substance abuse disorder, through the use of aversive agents or pharmacologic antagonists.
[0071] Pharmaceutical dosage forms containing abusable drugs have been used for non-medical purposes in a variety of settings: i) by patients with a disorder requiring treatment with an abusable drug who have developed an addiction disorder following initiation of therapy; ii) by patients with said disorder who had a pre-existing addiction disorder; iii) by patients with an addiction disorder seeking abusable drugs for their reinforcing, rewarding, euphoriant or other mood altering properties.
[0072] Non-medical users of abusable drugs are either recreational drug users who may use such agents episodically, or individuals with an addiction disorder who may require frequent maintenance doses. Abusable drugs may be ingested whole, crushed and ingested, crushed or vaporized and snorted or injected intravenously after attempted extraction of the active pharmaceutical ingredient.
[0073] Addicts and recreational drug users commonly use extended release versions of abusable drugs by a variety of routes of administration. Commonly used methods include 1) parenteral (e.g., intravenous injection), 2) intranasal (e.g., snorting), and 3) episodic or repeated oral ingestion of intact or crushed tablets or capsules.
[0074] One mode of abuse can involve the extraction of the abusable drugs component from the dosage form by first mixing the tablet or capsule with a suitable solvent (e.g., water or alcohol), and then filtering and/or extracting the abusable drugs component from the mixture for intravenous injection. Another mode of abuse of extended release abusable drugs can involve dissolving the drug in water, alcohol or another "recreational solvent" to hasten its release and to ingest the contents orally, in order to provide high peak concentrations and maximum euphoriant effects.
[0075] A number of strategies have been introduced to minimize the abuse of mood altering drugs. Primary among these schemes is a legal infrastructure that controls the manufacture, distribution and sale of such drugs. In the United States, the vast majority of abusable drugs having clinically useful effects are restricted to dispensing on a prescription-only basis. Most of these drugs are "scheduled" as "controlled drugs", such that distribution of the drug is subject to strict controls and overview. The idea behind scheduling abusable drugs as "controlled" is to ensure that the drugs are dispensed only for the amelioration of legitimate therapeutic maladies, and not for any mood-altering effect "high" or euphoria that may be produced by the drug when used in supra- therapeutic doses or administered by non-approved routes of administration.
[0076] While the scheduling of abusable drugs as "controlled drugs" has reduced abuse of the drugs, it has not been entirely successful. For example, some persons who are legitimately prescribed the drugs sometimes divert the drugs to persons seeking their procurement for "recreational uses." These "recreational drug users" are frequently willing to pay significant sums of money for the drugs. In other cases, certain health professionals, unfortunately, have been found to be culprits in the non-approved distribution of abusable drugs.
[0077] It is believed that the most widely used diversion techniques at the
"street level" are "doctor shopping" and prescription forgeries. In the case of the former, individuals who may or may not have a legitimate ailment requiring a doctor's prescription for controlled substances, visit numerous doctors, sometimes in several states, to acquire large amounts of controlled substances they abuse or sell to others. [0078] There is a growing recognition in the medical community that a large number of patients suffer from the undertreatment of their medical condition when the treatment involves the use of psychoactive drugs, particularly those drugs which tend to diverted and abused. Scheduling of abusable drugs has also had the unintentional side-effect of causing physicians, fearful of being accused of permitting or even promoting drug abuse and drug overuse, to prescribe suboptimal doses of abusable drugs to patients in need of them, and to prescribe less effective drugs to patients that are not similarly scheduled.
[0079] An additional issue with extended release forms of drugs, including abusable drugs is the interaction of the drug, even in an untampered form, r when consumed with alcohol. Under such conditions, a number of drugs have demonstrated an in vitro and in vivo propensity for significant dose dumping when they are co-ingested for medical purposes at prescribed doses with alcohol, increasing the potential for drug toxicity and further exacerbating the intensity of the (abusable) drug-alcohol pharmacodynamic interaction.
[0080] There is therefore also need, therefore, for novel methods of preventing excessive peak concentrations (dose dumping) of abusable drugs when they are co-ingested for medical purposes at prescribed doses with alcohol.
[0081] The present invention also involves oral pharmaceutical compositions of abusable drugs, including in extended release form and methods of use thereof which provide reduced variability in rate and extent of absorption when taken with food, compared with the fasted state.
[0082] Most pharmaceutical companies strive to develop oral pharmaceutical products which can be taken without regard to meal intake, i.e., on an empty stomach or with food. Indeed, it is now a requirement of both U.S. and E.U regulatory guidance's to conduct studies of all new chemical entities to determine the influence of concurrent food intake on the bioavailability of products. Concurrent intake of food in so called "fed- fasted" human bioavailability studies may increase, decrease or have no effect on the bioavailability of the pharmaceutical products. Accordingly, the prescribing information guides both the prescriber and the patient on the appropriate use of the pharmaceutical product.
[0083] The issue of food effects on oral bioavailability has particular importance with oral extended release products, due to the possibility of "dose dumping" where the is potential that a portion or a substantial portion of the dose intended to be released gradually over time (e.g., over 8, 12 or 24 hours) may be released instantaneously or rapidly, such that the peak plasma concentration of the drug (peak exposure) will be substantially increased, resulting in toxicity and the duration of effect will be significantly reduced, potentially resulting in reduced duration of therapeutic effect. The ability to take the drug without regard to food intake and in the case of extended release pharmaceutical products, the absence of dose dumping has been found to be a significant patient benefit and a competitive marketing advantage. It is not uncommon to see pharmaceutical advertising targeted to medical practitioners which states "no food effect", "may be taken on an empty stomach or with food" and "may be taken without regard to food" and "no change in bioavailability with food".
[0084] The U.S. prescribing information for OxyContin™ (oxycodone ER) states "Food has no significant effect on the extent of absorption of oxycodone from OxyContin. However, the peak plasma concentration of oxycodone increased by 25% when a OxyContin 160 mg Tablet was administered with a high-fat meal."
[0085] The U.S. prescribing information for Opana™ ER (oxymorphone ER) states "two studies examined the effect of food on the bioavailability of single doses of 20 and 40 mg of OPANA ER in healthy volunteers. In both studies, after the administration of OPANA ER, the Cmax was increased by approximately 50% in fed subjects compared to fasted subjects. A similar increase in Cmax was also observed with oxymorphone solution."
[0086] The U.S. prescribing information for Ritalin LA™ (methylphenidate
ER) states "When Ritalin LA was administered with a high fat breakfast to adults, Ritalin LA had a longer lag time until absorption began and variable delays in the time until the first peak concentration, the time until the interpeak minimum, and the time until the second peak. The first peak concentration and the extent of absorption were unchanged after food relative to the fasting state, although the second peak was approximately 25% lower. The effect of a high fat lunch was not examined".
[0087] The U.K. prescribing information for Equasym XL™
(methylphenidate ER) states: "Ingestion together with food with a high fat content delays its absorption (Tmax) by approximately one hour and increases the maximum concentration (Cmaχ) by approximately 30% and the amount absorbed (AUC) by approximately 17%."
[0088] The U.S. prescribing information for Aderall XR ™ (amphetamine and dextroamphetamine ER) states "Food does not affect the extent of absorption of d-amphetamine and 1- amphetamine, but prolongs T max by 2.5 hours (from 5.2 hrs at fasted state to 7.7 hrs after a high-fat meal) for d-amphetamine and 2.1 hours (from 5.6 hrs at fasted state to 7.7 hrs after a high fat meal) for 1- amphetamine after administration of ADDERALL XR M 30 mg."
[0089] In summary, many abusable drugs in extended release form have a potentially clinically important food effect.
[0090] There is therefore a need for oral immediate release pharmaceutical compositions, and particularly extended release pharmaceutical compositions with consistent bioavailability regardless of administration in the fed or fasted state.
[0091] There is a need for methods and pharmaceutical compositions of pharmaceuticals and therapeutic agents that provide an extended release profile, preferably suitable for every 8, 12 or 24 hour oral administration. There is a need for methods and pharmaceutical compositions of abusable drugs that provide an extended release profile. There is also a need for methods and pharmaceutical compositions of abusable drugs that provide an abuse deterrent profile. In particular, there is a need for methods and pharmaceutical compositions of abusable drugs that simultaneously provide abuse deterrence properties and extended release profiles, preferably suitable for every 8, 12 or 24 hour oral administration. There is also a need for extended release formulations of pharmaceuticals and therapeutic agents, and abusable drugs that are stable (i.e., do not dose dump) when used at therapeutic doses for medical purposes in conjunction with alcohol. There is also a need for extended release formulations of pharmaceuticals and therapeutic agents, and abusable drugs that provide a extended release pharmacokinetic profile suitable for every 8, 12 or 24 hour release and will be resistant to crushing at room temperature and upon freezing, melting to allow for filtration and/or aspiration into a syringe and extraction with recreational solvents. There is also a need for extended release formulations of pharmaceuticals and therapeutic agents, and abusable drugs that provide a extended release pharmacokinetic profile suitable for every 8, 12 or 24 hour release and will be resistant to crushing at room temperature and upon freezing, melting to allow for filtration and/or aspiration into a syringe and extraction with recreational solvents, all without doing harm to patients or patients with a substance abuse disorder, through the use of aversive agents or antagonists.
DETAILED DESCRIPTION OF THE INVENTION
[0092] In some preferred embodiments, the present invention is directed at pharmaceutical compositions of abusable drugs to provide abuse deterrence properties.
[0093] hi some preferred embodiments, the present invention is directed at pharmaceutical compositions of abusable drugs to provide extended release properties.
[0094] In some preferred embodiments, the present invention is directed at pharmaceutical compositions of abusable drugs that provide simultaneous abuse deterrence properties and extended release properties.
[0095] In some preferred embodiments, the present invention is directed at pharmaceutical compositions of abusable drugs that provide simultaneous abuse deterrence properties and extended release properties using substantially the same ingredients to achieve abuse deterrence properties and extended release.
[0096] In some preferred embodiments, the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide abuse deterrence properties.
[0097] In some preferred embodiments, the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide extended release properties.
[0098] In some preferred embodiments, the present invention is directed at oral solid pharmaceutical compositions of abusable drugs that are in the form of a liquid, semisolid, oil or otherwise difficult to granulate.
[0099] In some preferred embodiments, the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide simultaneous abuse deterrence properties and extended release properties.
[00100] In some preferred embodiments, the present invention is directed at liquid pharmaceutical compositions of abusable drugs that solidify at room temperature to provide simultaneous abuse deterrence properties and extended release properties using substantially the same ingredients to achieve abuse deterrence properties and extended release.
[00101] In some preferred embodiments, the present invention is directed at oral abusable drug pharmaceutical compositions and the use thereof for preventing or minimizing the risk of abusable drug toxicity from either intentional or unintentional tampering.
[00102] In some preferred embodiments, the present invention is directed at oral abusable drug pharmaceutical compositions and the use thereof for deterring abuse by drug addicts and/or recreational drug users.
[00103] In some preferred embodiments, the present invention is directed at oral abusable drug pharmaceutical compositions that provide extended release delivery of the drug and the use thereof for the treatment of pain, addiction disorders, spasticity, musculoskeletal disorders, ADHD, insomnia, excessive sleepiness, daytime sleepiness, sleep disorders, anxiety disorders, panic attacks, agoraphobia, obsessive-compulsive disorders, psychiatric disorders, neurologic disorders, excess weight, obesity and other medical maladies responsive to treatment with the abusable drugs.
[00104] Representative example of abusable drugs are, without limitation amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, nicotine, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof, but specifically excluding all opioid agonists.
[00105] As used herein, "abusable drugs", "abusable drug", "abusable pharmaceuticals", "abusable substances" and "abusable dosage forms" are: (i) substances which have medical applications for the prevention or treatment of diseases and disorders (i.e., "therapeutically active agents"); and (ii) substances which have the potential for being abused by drug abusers, recreational drug users and individuals with an addiction disorder for intermittent use, recreational use or chronic use, wherein the abusable drug is being abused for one or more of the following effects: mood alterations; euphoria, pleasure; a feeling of high; a feeling of drug liking; anxiolysis; mental stimulation; increased mental arousal; sedation; calmness; a state of relaxation; psychotomimesis; hallucinations; alterations in perception, cognition and mental focus; insomnia; hypersomnia; increased wakefulness or alertness; memory improvement; increased sexual gratification; increased sexual arousal; increased sexual desire and sexual anticipation; increased socialization; reduced social anxiety; psychologically reinforcement; and psychologically rewarding. It reasons that various abusable drugs of the invention will have different abusable characteristics being sought after by the drug addict or recreational drug user. Abusable drugs of the invention specifically exclude all opioid agonists.
[00106] In some preferred embodiments, abusable drugs of the present invention can be formulated with the substantially the same ingredients to deter abuse and minimize abusable drug toxicity on tampering while simultaneously providing an extended release pharmacokinetic profile suitable for every 4, 6, 8, 12 or 24 hour dosing.
[00107] In some preferred embodiments, abusable drugs of the present invention can be formulated with the substantially the same ingredients to deter abuse and minimize abusable drug toxicity on tampering while simultaneously providing an extended release pharmacokinetic profile suitable for every 4, 6, 8, 12 or 24 hour dosing, without the need to include an aversive agent or an antagonist for the abusable drug in the formulation.
[OOIOS] In some preferred embodiments, abusable drug pharmaceutical compositions and methods of the present invention provide (i) abuse deterrence; (ii) extended release; and (iii) simultaneous abuse deterrence and extended release, prepared using one or more ADER compounds.
[00109] In some preferred embodiments, abusable drug pharmaceutical compositions and methods of the present invention provide simultaneous abuse deterrence and extended release, prepared using ADER, using substantially the same ingredients to effect abuse deterrence and extended release.
[00110] In some preferred embodiments, abusable drug pharmaceutical compositions and methods of the present invention provide (i) extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER.
[00111] In some preferred embodiments, abusable drug pharmaceutical compositions and methods of the present invention provide (i) extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER, using substantially the same ingredients to effect extended release and protection against ethanol induced dose dumping. [00112] In some preferred embodiments, abusable drug pharmaceutical compositions and methods of the present invention provide (i) abuse deterrence and extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER.
[00113] In some preferred embodiments, abusable drug pharmaceutical compositions and methods of the present invention provide (i) abuse deterrence and extended release; and (ii) protection against ethanol induced dose dumping, prepared using ADER, using substantially the same ingredients to effect abuse deterrence and extended release, and protection against ethanol induced dose dumping.
[00114] In some preferred embodiments, the present invention is directed to a novel method for reducing the peak concentration (Cmaχ) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
[00115] In some preferred embodiments, the present invention is directed to a novel method for reducing the early post-dose partial area under the plasma concentration time curve (e.g., AUC0.2, AUCo-4 and AUCo-6) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
100116] In some preferred embodiments, the present invention is directed to a novel method for reducing the early post-dose average plasma concentration time (Cave) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
[00117] In some preferred embodiments, the present invention is directed to a novel method for reducing the incidence of abusable drug toxicity upon tampering of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
[00118] In some preferred embodiments, the present invention is directed to a novel method for reducing the intensity of abusable drug toxicity upon tampering of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER. [00119] In some preferred embodiments, the present invention is directed to a novel method for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including nausea, vomiting, somnolence, stupor, coma, respiratory depression, apnea, respiratory arrest, circulatory depression, bradycardia, hypotension, shock and skeletal muscle flaccidity, said method comprising administering the abusable drug and a suitable amount of ADER.
[00120] hi some preferred embodiments, the present invention is directed to a novel method for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including tachycardia, mood alteration, euphoria, CNS stimulation, agitation, increased sweating, psychotomimetic effects, hallucinations, perception alterations, cognitive alterations, reinforcing effects and pleasurable effects said method comprising administering the abusable drug and a suitable amount of ADER.
[00121] hi some preferred embodiments, the present invention is directed to a novel method for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including "high", "liking", pleasurable, euphoric, alertness, wakefulness, calming, anxiolytic, auditory and visual perceptual alterations, relaxing, analgesic and rewarding effects.
[00122] In some preferred embodiments, the present invention, is directed to novel pharmaceutical compositions for use in reducing the peak concentration (Cmax) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
[00123] In some preferred embodiments, the present invention is directed to novel pharmaceutical compositions for reducing the early post-dose partial area under the plasma concentration time curve (e.g., AUCo-25 AUCo-4 and AUC0-6) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER.
[00124] . In some preferred embodiments, the present invention is directed to novel pharmaceutical compositions for reducing the early post-dose average plasma concentration time (Cave) of the abusable drug, said method comprising administering the abusable drug and a suitable amount of ADER. [00125] In some preferred embodiments, the present invention is directed to novel pharmaceutical compositions for reducing the incidence of abusable drug toxicity, said method comprising administering the abusable drug and a suitable amount of ADER.
[00126] In some preferred embodiments, the present invention is directed to novel pharmaceutical compositions for reducing the intensity of abusable drug toxicity, said method comprising administering the abusable drug and a suitable amount of ADER.
[00127] In some embodiments, the invention provides an oral pharmaceutical composition of an abusable oral drug which is: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and (e) resistant to intentional or surreptitious adulteration of beverages; or two or more of the above [(a) to (e)], said composition comprising: (A) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof; and (B) One or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes.
[00128] In some embodiments, the invention provides an oral pharmaceutical composition of an abusable oral drug which is: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and (e) resistant to intentional or surreptitious adulteration of beverages; or two or more of the above [(a) to (e)], said composition comprising: (A) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof; and (B) Two or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes.
[00129] In some embodiments, the invention provides an oral pharmaceutical composition of an abusable oral drug which is: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and (e) resistant to intentional or surreptitious adulteration of beverages; or two or more of the above [(a) to (e)], said composition comprising: (A) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof; and (B) Two or more compounds selected from at least two categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes.
[00130] In some embodiments, the invention provides an oral pharmaceutical composition of an abusable oral drug which is: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and (e) resistant to intentional or surreptitious adulteration of beverages; or two or more of the above [(a) to (e)], said composition comprising: (A) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof; and (B) One or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said composition [(a) to (e)] using substantially the same ingredients.
[00131] In some embodiments, the invention provides an oral pharmaceutical composition of an abusable oral drug which is: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and (e) resistant to intentional or surreptitious adulteration of beverages; or two or more of the above [(a) to (e)], said composition comprising: (A) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof; and (B) Two or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said composition [(a) to (e)] using substantially the same ingredients.
[00132] In some embodiments, the invention provides an oral pharmaceutical composition of an abusable oral drug which is: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and (e) resistant to intentional or surreptitious adulteration of beverages; or two or more of the above [(a) to (e)], said composition comprising: (A) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof; and (B) Two or more compounds selected from at least two categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said composition [(a) to (e)] using substantially the same ingredients.
[00133] In some embodiments, compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides for abuse deterrence of the abusable drugs.
[00134] In some embodiments, compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form resists, deters or prevents crushing, shearing, grinding, chewing, dissolving, melting, needle aspiration, inhalation, insufflation or solvent extraction of the abusable drug.
[00135] In some embodiments, compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides extended release of the abusable drug.
[00136] In some embodiments, compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g:, glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides abuse deterrence and extended release of the abusable drug.
[00137] In some embodiments, compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides simultaneous abuse deterrence and extended release of the abusable drug.
[00138] In some embodiments, compositions and methods of the present invention include: (i) one or more abusable drugs (e.g., amphetamine and amphetamine like CNS-stimulants, methylphenidate, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates or cannabinoid agonists); and (ii) ADER; and (iii) optionally, other therapeutic agents in immediate or extended release form; and (iv) optionally one or more excipients or auxiliary agents (e.g., glidants, lubricants, disintegrants, antistatic agents, solvents, channel forming agents, coating agents, flavorants, preservatives, bulking agents, polymers, etc) and inert carriers; wherein the dosage form provides simultaneous abuse deterrence and extended release of the abusable drug using substantially the same ingredients.
[00139] In some preferred embodiments, the present invention is directed to novel pharmaceutical compositions for reducing the intensity or frequency of one or more symptoms, including nausea, vomiting, somnolence, stupor, coma, respiratory depression, apnea, respiratory arrest, circulatory depression, bradycardia, hypotension, shock and skeletal muscle flaccidity, said method comprising administering the abusable drug and a suitable amount of ADER.
[00140] In some preferred embodiments, the present invention is directed to novel pharmaceutical compositions for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including tachycardia, mood alteration, euphoria, CNS stimulation, agitation, increased sweating, psychotomimetic effects, hallucinations, perception alterations, cognitive alterations, reinforcing effects and pleasurable effects said method comprising administering the abusable drug and a suitable amount of ADER.
[00141] In some preferred embodiments, the present invention is directed to a novel pharmaceutical compositions for reducing the intensity or frequency of one or more signs and symptoms of abusable drug toxicity, including "high"; "liking"; "pleasurable"; "euphoric"; "alertness"; "wakefulness"; "calming"; "anxiolytic"; auditory and visual perceptual alterations; "relaxing" and "rewarding" effects.
[00142] In some preferred embodiments, the present invention is directed to a novel method and pharmaceutical compositions for preventing or minimizing excessive peak concentrations (dose dumping) of therapeutic doses of extended release abusable drugs used for medical purposes, when they are co- ingested with alcohol.
[00143] In some preferred embodiments, the present invention is directed to a novel method and pharmaceutical compositions for reducing the solvent extraction efficiency of the dosage form upon tampering.
[00144] In some preferred embodiments, the present invention is directed to a novel method and pharmaceutical compositions for reducing the filtration efficiency of the dosage form upon tampering.
[00145] In some preferred embodiments, the present invention is directed to a novel method and pharmaceutical compositions for preventing the surreptitious adulteration of beverages.
[00146] In some preferred embodiments, the present invention is directed . pharmaceutical compositions which include one or more abusable drugs alone or in combination with other therapeutic agents, one or more ADER agents specified herein, and optionally one or more excipients (e.g., glidants, lubricants, disintegrants, etc) and inert carriers, said composition resisting, deterring, discouraging or preventing crushing, shearing, grinding, chewing, dissolving, melting, needle aspiration, inhalation, insufflation, solvent extraction and filtration of the abusable drug.
[00147] In some preferred embodiments, pharmaceutical compositions of the present invention provide a more extended release pharmacokinetic profile compared with formulations devoid of ADER.
[00148] In some preferred embodiments, pharmaceutical compositions and methods of the present invention can form a viscous substance upon contact with a solvent such that the abusable drug cannot be easily drawn into a syringe; crushed and powdered to facilitate or enhance nasal delivery (snorting or nasal insufflation), inhalation or rapid oral delivery of a larger than medically intended delivery of the abusable drug; extracted with solvents and filtered.
[00149] In some preferred embodiments, the pharmaceutical composition resists the rapid release of all or substantially all of the abusable drug content of the unit dose upon tampering. In another preferred embodiment of the invention, the pharmaceutical composition resists the rapid release of a portion of the abusable drug content of the unit dose upon tampering. In yet another preferred embodiment of the invention, upon tampering, the abusable drug formulated with ADER resists the release of the abusable drug to a greater extent than when formulated without ADER.
[00150] In some preferred embodiments, the pharmaceutical composition resists the rapid release of all or substantially all of the abusable drug content of the unit dose upon co-administering with alcohol. In another preferred embodiment of the invention, the pharmaceutical composition resists the rapid release of a portion of the abusable drug content of the unit dose upon coadministering with alcohol. In yet another preferred embodiment of the invention, upon co-administering with alcohol, the abusable drug formulated with ADER resists the release of the abusable drug to a greater extent than when formulated without ADER.
[00151] In some preferred- embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be filled in a hard gelatin capsule without banding. In some preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be filled in a hard gelatin capsule with security banding. Li another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be filled in a soft shell capsules. In another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be prepared as a solid dispersion for direct compression into tablets, for extrusion and pelletization, followed by compression into a tablet or filling into a capsule. In another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be compressed into tablets. In another preferred embodiment of the abuse deterrent pharmaceutical composition, the therapeutic pharmaceutical composition can be prepared into multiparticulate matrices followed by tabletting or capsule filling.
[00152] The present invention is directed at oral pharmaceutical compositions of abusable drugs or their pharmaceutically acceptable salts or mixtures thereof.
[00153] The present invention also relates to oral abusable drug pharmaceutical compositions and methods for the prevention and treatment of pain, musculoskeletal disorders, spasticity, ADHD, sleep disorders, anxiety, obsessive-compulsive disorders, psychiatric disorders, neurologic disorders, obesity and other medical conditions amenable to treatment with the abusable drug.
[00154] It is an object of certain preferred embodiments of the present invention to substantially improve the efficiency and quality of disease management for: (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug.
[00155] It is an . object of certain preferred embodiments of the present invention to provide bioavailable oral formulations of abusable drugs suitable for up to once-daily (e.g., Q4H, Q6H, Q8H, Q12H, Q24H) administration which substantially improve the efficiency and quality of disease management for: (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug. [00156] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral formulations of abusable drugs which provide a substantially increased duration of effect as compared to immediate release formulations.
[00157] ' It is an object of certain preferred embodiments of the present invention to provide bioavailable oral formulations of abusable drugs which provide a substantially reduced abuse potential compared with immediate release formulations of abusable drugs.
[00158] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral formulations of abusable drugs which provide a substantially reduced abuse potential compared with currently available extended release formulations.
[00159] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral formulations of abusable drugs which provide a substantially reduced abuse potential compared with commercially available formulations of abusable drugs.
[00160] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral immediate release formulations of abusable drugs which provide a substantially reduced abuse potential.
[00161] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral immediate release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with food, compared with the fasted state.
[00162] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral immediate release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with alcohol, compared to an alcohol-free state.
[00163] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral extended release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with food, compared with the fasted state. [00164] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral extended release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with alcohol, compared to an alcohol-free state.
[00165] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral abuse resistant and abuse deterrent extended release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with food, compared with the fasted state.
[00166] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral abuse resistant and abuse deterrent extended release formulations of abusable drugs which provide a substantially reduced variability in rate and extent of absorption when taken with alcohol, compared to an alcohol-free state.
[00167] It is an object of certain preferred embodiments of the present invention to provide bioavailable oral extended release formulations of abusable drugs which provide a substantially reduced abuse potential compared with currently available extended release formulations.
[00168] It is an object of certain preferred embodiments of the present invention to provide bioavailable formulations for oral administration suitable for up to once-a-day administration (e.g., Q4H, Q6H, Q8H, Ql 2H, and Q24H).
[00169] It is an object of certain preferred embodiments of the present invention to provide bioavailable formulations for oral administration suitable for up to once-a-day administration which provide an early onset and sustained duration of therapeutic effect.
[00170] It is an object of certain preferred embodiments of the present invention to provide formulations of abusable drugs which provide a therapeutic effect for up to about 30 minutes. In other preferred embodiments, the formulations of abusable drugs provide a therapeutic effect for up to about 1 hour, or up to about 2 hours, or up to about 4 hours, or up to about 6 hours, or up to about 8 hours, or up to about 10 hours, or up to about 12 hours, or up to about 16 hours, or up to about 18 hours, or up to about 24 hours or up to about 36 hours, or up to about 48 hours.
[00171] It is an object of certain preferred embodiments of the invention to provide a method and formulations of oral abusable drugs for the prevention and treatment of: (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug.
[00172] It is an object of certain preferred embodiments of the invention to provide a method and formulations of oral abusable drugs for the prevention and treatment of (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug; said formulations and methods not having a propensity of substantial drug accumulation.
[00173] It is an object of certain preferred embodiments of the invention to provide a method and formulations of oral abusable drugs for the prevention and treatment of (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug; said formulations having a reduced potential for tampering (e.g., mechanical, thermal, chemical or physical tampering). [00174] It is an object of certain preferred embodiments of the invention to provide a method and formulations of oral abusable drugs for the prevention and treatment of (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug; said formulations in some embodiments having a reduced potential for drug abuse (e.g., inhalational, intranasal, intravenous or oral abuse); said formulations in some embodiments having a reduced potential for drug diversion; said formulations in some embodiments having a reduced intrasubject and intrasubject pharmacokinetic variability; said formulations in some embodiments having a reduced intersubject and intrasubject pharmacodynamic variability; said formulations in some embodiments having a reduced peak to trough fluctuation; said formulations in some embodiments having a shorter time to therapeutic concentrations and a shorter time to steady-state; said formulations in some embodiments being in extended release dosage form, and said formulations providing an extended duration of action; said formulations in some embodiments providing more than one of the aforementioned properties.
[00175] It is an object of certain preferred embodiments of the invention to provide a method and formulations of oral abusable drugs for the prevention and treatment of pain, said formulations suitable for use in acute pain, including acute postsurgical pain. In other preferred embodiments, the invention provides a method and formulations of oral abusable drugs for the prevention and treatment of chronic pain, cancer pain, neuropathic pain, somatic pain, visceral pain, idiopathic pain and breakthrough pain of various etiologies, including cancer, chronic pain and neuropathic pain.
[00176] It is an object of certain preferred embodiments of the invention to provide a method and formulations of oral abusable drugs for the prevention and treatment of one or more of the following: ADHD; or obesity; or sleep disorders (.e., insomnia, hypersomnia, narcolepsy, excessive daytime sleepiness); or anxiety disorders (e.g., generalized anxiety, agoraphobia, panic attacks); or obsessive compulsive disorder; or nicotine addiction (e.g., through nicotine substitution or nicotine maintenance therapy); or psychiatric disorders; or neurologic disorders; or addiction disorders.
[00177] It is an object of certain embodiments of the present invention to provide oral formulations of abusable drugs with both immediate release and controlled release forms.
[00178] It is an object of certain embodiments of the present invention to provide oral formulations of abusable drugs in pulsatile release form.
[00179] It is an object of certain embodiments of the present invention to provide abusable drugs for oral administration wherein the abusable drugs are dispersed within a matrix.
[00180] In certain preferred embodiments the oral dosage form of the present invention comprises a matrix which includes ADER and an abusable drug or a pharmaceutically acceptable salt thereof. In certain preferred embodiments, the matrix is compressed into a tablet and may be optionally overcoated with a coating that in addition to the sustained release material of the matrix may control the release of the abusable drug or pharmaceutically acceptable salt thereof from the formulation, such that blood levels of active ingredient are maintained within the therapeutic range over an extended period of time. In certain alternate embodiments, the matrix is encapsulated.
[00181] In certain preferred embodiments, the sustained release oral dosage form of the present invention comprises ADER and a plurality of pharmaceutically acceptable sustained release matrices comprising an abusable drug or a pharmaceutically acceptable salt thereof, the dosage form maintaining the plasma levels of abusable drug within the therapeutic range over an extended period of time when administered to patients.
[00182] In some preferred embodiments of the invention, the abusable drugs are in a matrix that is in the form of pellets or beads. 100183] In some preferred embodiments, the dosage form of the invention comprises a compressed tablet, compressed capsule or uncompressed capsule. In other embodiments, the dosage form comprises a liquid fill capsule.
[00184] In some preferred embodiments, the dosage form of the invention comprises an oral formulation (e.g., tablet or capsule) which is coated to prevent substantial direct contact of abusable drug with oral cavity (e.g. tongue, oral mucosa), oropharyngeal mucosal surface, esophagus or stomach. In some preferred embodiments, the dosage form of the invention comprises an oral formulation which is coated with a film or polymer. In some preferred embodiments, the dosage form of the invention comprises abusable drugs in an enteric coating. In some preferred embodiments, the dosage form of the invention comprises abusable drugs formulated with pharmaceutical excipients and auxiliary agents known in the art, such that the abusable drug is released after a approximately specific amount of time, or at an approximately specific anatomic location in the gastrointestinal tract, or when the dosage form is in contact with specific gastrointestinal conditions (e.g., pH range, osmolality, electrolyte content, food content.).
[00185] In some preferred embodiments, the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined under fed conditions. In other preferred embodiments, the in vivo pharmacokinetic parameters are derived or determined under fasted conditions.
[00186] Some or all of the above objects and other objects herein are achieved by embodiments of the present invention, which is directed in part to a dosage form of oral abusable drugs in abuse deterrent form.
[00187] Some or all of the above objects and other objects herein are achieved by embodiments of the present invention, which is directed in part to a dosage form of oral abusable drugs in extended release form.
[00188] Some or all of the above objects and other objects herein are achieved by embodiments of the present invention, which is directed in part to a dosage form of oral abusable drugs in an abuse deterrent and extended release form. [00189] Some or all of the above objects and other objects herein are achieved by embodiments of the present invention, which is directed in part to a dosage form of oral abusable drugs and ADER. [00190] Some or all of the above objects and other objects herein are achieved by embodiments of the present invention, which is directed in part to a dosage form of oral extended release abusable drugs and ADER. [00191] Some or all of the above objects and other objects herein are achieved by embodiments of the present invention, which is directed in part to a dosage form of abuse deterrent abusable drugs and ADER. [00192] Some or all of the above objects and other objects herein are achieved by embodiments of the present invention, which is directed in part to a dosage form which provides simultaneous abuse deterrence and extended release through the inclusion of ADER. [00193] The embodiments, compositions and methods of the present invention specifically exclude opioids, opioid agonists and opioid analgesics. [00194] In some preferred embodiments, compositions and methods of the present invention exclude the prevention or treatment of pain. [00195] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs and ADER, said compositions and methods excluding all drugs for the management of pain. [00196] As defined herein, the term "abusable drugs" specifically excludes all opioid agonists. [00197] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS -stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; and (iv) cannabinoid agonists. [00198] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; and (iv) cannabinoid agonists.
[00199] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; and (iv) cannabinoid agonists.
[00200] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances which are scheduled drugs under the United States Controlled Substances Act of 1970, as amended.
[00201] As used herein, the phrase "scheduled drugs under the United States
Controlled Substances Act of 1970" and "United States Controlled Substances Act of 1970" specifically excludes opioid agonists.
[00202] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended); (vi) drugs listed under the United States Psychotropic Substances Act of 1978, as amended.
[00203] As used herein, the phrase "drugs listed under the United States
Psychotropic Substances Act of 1978" and "the United States Psychotropic Substances Act of 1978" specifically excludes opioid agonists.
[00204] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended).
[00205] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include: (i) amphetamine and amphetamine like CNS-stimulants; (ii) methylphenidate; (iii) benzodiazepine agonists and non-benzodiazepine hypnotics and CNS depressants; (iv) barbiturates; (iv) cannabinoid agonists; and (v) controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended); (vi) drugs listed under the United States Psychotropic Substances Act of 1978, as amended.
[00206] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof.
[00207] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs include amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics, nicotine, nicotine analogs, tobacco extract, and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof. [00208] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to amphetamine and amphetamine like CNS-stimulants. [00209] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to methylphenidate, methylphenidate prodrugs and methylphenidate esters. [00210] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to benzodiazepine agonists [00211] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to barbiturates. [00212] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to cannabinoid agonists. [00213] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to prodrugs of abusable drugs. [00214] In some preferred embodiments, compositions and methods of the present invention involve abusable drugs, wherein the abusable drugs are limited to nicotine. [00215] In some preferred embodiments, the dosage form is a capsule, said capsule rendered tamper-resistant with a security banding seal between the capsule parts. [00216] In some preferred embodiments, the dosage form is a capsule, said capsule rendered liquid tight or leak resistant with a banding seal between the capsule parts. [00217] In another aspect, the invention relates to a method and pharmaceutical compositions for prevention or treatment of (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive- compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug; comprising oral administration of a dosage form • containing an abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER. 8] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition for the prevention and treatment of (i) pain; (ii) ADHD; (iii) insomnia; (iv) excessive sleepiness; (v) daytime sleepiness; (vi) generalized anxiety; (vii) agoraphobia; (viii) panic disorders; (ix) obesity; (x) obsessive-compulsive disorders; (xi) spasticity; (xii) musculoskeletal disorders; (xiii) sleep disorders; (xiv) psychiatric disorders; (xv) neurologic disorders; (xvi) addiction disorders; or (xvii) other medical conditions amenable to treatment with the abusable drug; comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form: (a) abuse deterrent; and/or (b) extended release; and/or (c) resistant to significant alcohol dose-dumping; and/or (d) resistant to significant variations in rate and/or extent of absorption based on a fed or fasted state; and/or (e) resistant to intentional or unintentional tampering; and/or (f) resistant to abusable drug toxicity due to intentional or unintentional tampering; and/or (g) resistant to significant variations in clinical effects when ingested after attempted tampering; and/or (h) resistant to intentional or surreptitious adulteration of beverage; and/or (i) resistant to significant drug-food interaction (i.e., large changes in rate or extent of absorption when taken with food); said rendering by said ADER material achieved in some embodiments using substantially the same ADER material; said dosage form in some embodiments suitable for administration up to about every 2 hours, or up to about every 4 hours, or up to about every 6 hours, or up to about every 8 hours, or up to about every 12 hours, or up to about every 24 hours to a human patient; said dosage form in some embodiments having a therapeutic effect of up to about every 1 hour, or up to about every 4 hours, or up to about every 6 hours, or up to about every 8 hours, or up to about every 12 hours, or up to about every 24 hours upon administration to a human patient.
[00219] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form abuse deterrent, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency, hi other preferred embodiments, the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
[00220] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug .or a mixture thereof and ADER material to render said dosage form extended release; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency, hi other preferred embodiments, the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency. [00221] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form abuse deterrent and extended release, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency. In other preferred embodiments, the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
[00222] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form resistant to alcohol dose dumping, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency. In other preferred embodiments, the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
[00223] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug or a mixture thereof and ADER material to render said dosage form resistant to significant fluctuations in bioavailability when given under fed and fasted conditions, said dosage form suitable for up to every 24 hour (once-a-day) administration to a human patient; said dosage form providing at least 60% of the steady state concentration of abusable drug after administration of one dose at its intended dosing frequency. In other preferred embodiments, the dosage form provides at least about 62.5%, or at least about 65%, or at least about 67.5%, or at least about 70%, or at least about 72.5%,or at least about 75%, or at least about 77.5%, or at least about 80%, or at least about 82.5%,or at least about 85%, or at least about 87.5%, or at least about 90%, or at least about 92.5%, or at least about 95% or at least 98% of the steady state therapeutic concentration of abusable drug after administration of one dose at its intended dosing frequency.
[00224] In some preferred embodiments, the invention comprises an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or a pharmaceutically acceptable salt of abusable drug, or a mixture thereof and ADER to render said dosage form suitable for three times a day administration (TID) or about every eight hours administration (Q8H).
[00225] In some preferred embodiments, the TED or Q8H oral pharmaceutical composition of an abusable drug provides a therapeutic effect for about 8 hours.
[00226] In some preferred embodiments, the TDD or Q8H oral pharmaceutical composition of an abusable drug provides a Cmaχ of abusable drugs at about 1 to about 6 hours.
[00227] In some preferred embodiments, the TID or Q8H oral pharmaceutical composition of abusable drugs provide a Cmin of abusable drugs at about 6 to 10 hours.
[00228] In some preferred embodiments, the TID or Q8H oral pharmaceutical composition of abusable drugs provide a mean abusable drugs Cs/Cmax ratio of 0.25 to about 0.95. [00229] In some preferred embodiments, the TDD or Q8H oral pharmaceutical composition of abusable drugs provide a percent fluctuation of less than 400%.
[00230] In some preferred embodiments, the TED or Q8H oral pharmaceutical composition of an abusable drug provides a W50 of 1.5 to about 6.5 hours.
[00231] In some preferred embodiments, the TDD or Q8H oral pharmaceutical composition of an abusable drug provides an HVD of 2 to about 7 hours.
[00232] In some preferred embodiments, the TDD or Q8H oral pharmaceutical composition of an abusable drug provides an HVD of about 2 to about 7 hours.
[00233] In some preferred embodiments, the TDD or Q8H oral pharmaceutical composition of an abusable drug an AI of not more that 4.0.
[00234] In some preferred embodiments, the invention comprises an oral pharmaceutical composition comprising therapeutically effective amounts of abusable drug or pharmaceutically acceptable salts thereof, or mixtures thereof and ADER; said dosage from providing a Cmax of abusable drug occurring from a mean of about 0.25 to about 30 hours. In other preferred embodiments, the dosage form provides a Cmaχ of abusable drug occurring from a mean of about 0.5 to about 30 hours, or from a mean of about 1 to about 30 hours, or about 1 to about 26 hours, or about 1 to about 24 hours, or about 1 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours, or about 1 to about 14 hours, or about 1 to about 12 hours, or about 1 to about 10 hours, or about 1 to about 8 hours, or about 1 to about 6 hours, or about 1 to about 4 hours, or about 1 to about 3 hours, or about 2 to about 30 hours, or about 4 to about 30 hours, or about 4 to about 24 hours, or about 6 to about 24 hours, or about 8 to about 24 hours, or about 10 to about 20 hours, or about 12 to about 24 hours, or about 18 to about 24 hours, or about 2 to about 12 hours, or about 3 to about 12 hours, or about 3 to about 8 hours, or about 4 to about 10 hours, or about 4 to about 12 hours, or about 4 to about 9 hours, or about 5 to about 8 hours.
[00235] In some preferred embodiments, the invention comprises an oral pharmaceutical composition comprising therapeutically effective amounts of abusable drug or pharmaceutically acceptable salts thereof, or mixtures thereof and ADER; said dosage from providing a Cmjn of abusable drug occurring from a mean of about 0.5 to about 28 hours, or about 1 to about 28 hours, or about 1 to 24 hours, or about 1 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours, or about 1 to about 12 hours, or about 1 to 10 hours, or about 1 to about 8 hours, or about 1 to about 6 hours, or about I to about 4 hours, about 2 to about 24 hours, or about 3 to 24 hours, or about 4 to about 24 hours, or about 6 to about 24 hours, or about 8 to about 24 hours, about 2 to about 12 hours, or about 3 to 10 hours, or about 3 to about 8 hours, or about 4 to about 8 hours, or about 6 to about 10 hours.
[00236] In some preferred embodiments, the invention comprises an oral pharmaceutical composition comprising therapeutically effective amounts of abusable drug or pharmaceutically acceptable salts thereof, or mixtures thereof and ADER; said dosage form providing a systemic exposure as assessed by the mean abusable drug area under the plasma concentration time curve (AUCo-O after first administration which is at least about 40% of the area under the plasma drug concentration-time curve from time zero to infinity (AUCo-). In other preferred embodiments, the dosage from provides an AUCo-t which is at least about 45%, or which is at least about 50%, or which is at least about 55%, or at least about 60%, or which is at least about 65%, or at least about 70%, or which is at least about 75%, or at least about 80%, or at least about 85%, or at least about 88%, or at least about 90%, or at least about 92%, or at least about 94%, or at least about 96% or at least about 98% of the AUC0-.,.
100237] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of an abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form providing at least 80% of the steady state therapeutic concentration of abusable drug after administration of < three doses at their intended dosing frequency. In other preferred embodiments, said dosage form provides at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 85%, or at least about 90%, or at least about 92%, or at least about 95%, or at least about 97%, or at least about 99% of the steady state therapeutic concentration of abusable drug after administration of < three doses at their intended dosing frequency.
[00238] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form providing at least 80% of the steady state therapeutic concentration of abusable drug after administration of < two doses at their intended dosing frequency. In other preferred embodiments, said dosage form provides at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 85%, or at least about 90%, or at least about 92%, or at least about 95%, or at least about 97%, or at least about 99% of the steady state therapeutic concentration of abusable drug after administration of < two doses at their intended dosing frequency.
[00239] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form providing at least 80% of the steady state therapeutic concentration of abusable drug after administration of one dose at their intended dosing frequency. In other preferred embodiments, said dosage form provides at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 85%, or at least about 90%, or at least about 92%, or at least about 95%, or at least about 97%, or at least about 99% of the steady state therapeutic concentration of abusable drug after administration of one dose at their intended dosing frequency.
[00240] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing a Cmϊn/Cmax ratio of abusable drug of 0.1 to about 1.0. In other preferred embodiments, the dosage form provides a Cmin/Cmax ratio of abusable drug of about 0.1 to about 0.9, or about 0.1 to about 0.8, or about 0.1 to about 0.7, or about 0.1 to about 0.6, or about 0.1 to about 0.5, or about 0.1 to about 0.4, or about 0.1 to about 0.3, or about 0.2 to about 1.0, or about 0.25 to about 1.0, or about 0.4 to about 1.0, or about 0.5 to about 0.9, or about 0.5 to about 0.85, or about 0.5 to about 0.8, or about 0.5 to about 0.75, or about 0.5 to about 1.0, or about 0.65 to about 1.0, or about 0.75 to about 1.0, or about 0.2 to about 0.9, or about 0.3 to about 0.95, or about 0.3 to about 0.85,or about 0.3 to about 0.8, or about 03 to about 0.75, or about 0.3 to about 0.7, or about 0.3 to about 0.6, or about 0.4 to about 0.9, or about 0.4 to about 0.8, or about 0.4 to about 0.7, or about 0.4 to about 0.6, or about 0.8 to about 1, or about 0.8 to about 1.1.
[00241] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing a percent fluctuation of abusable drug of less than 400%. In other preferred embodiments, the dosage form provides a percent fluctuation of abusable drug of less than 350%, or less than 300%, or less than 250%, or less than 200%, or less than 150%, or less than 100%, or less than 75%, or less than 50%, or less than 25%.
[00242] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing a W50 of abusable drug of about 1 to about 6 hours for each 6 hour time period of intended dosing frequency and intended duration of action. In other preferred embodiments, the dosage form provides a W50 of abusable drug for each 6 hour time period of intended dosing frequency and intended duration of action of about 1 to about 5 hours, or about 1 to about 4 hours, or about 1 to about 3 hours, or about 1 to about 2 hours, or 2 to about 6 hours, or about 3 to about 6 hours, or about 4 to about 6 hours, or about 2 to about 4 hours. [00243] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing an HVD of abusable drug of about 1.5 to about 6 hours for each 6 hour time period of intended dosing frequency and intended duration of action. In other preferred embodiments, the dosage form provides a HVD of abusable drug for each 6 hour time period of intended dosing frequency and intended duration of action of about 1.5 to about 5 hours, or about 1.5 to about 4 hours, or about 1.5 to about 3 hours, or about 1.5 to about 2 hours, or 2 to about 6 hours, or about 3 to about 6 hours, or about 4 to about 6 hours, or about 2 to about 4 hours.
[00244J In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage form after administration to a human patient providing an AI of abusable drug of not more than 3.0. In other preferred embodiments, the dosage form provides an AI of abusable drug of not more than about 2.5, or not more than about 2, or not more than about 1.75, or not more than about 1.5, or not more than about 1.25, or not more than about 1, or not more than about 0.75, or not more than about 0.5, or not more than about 0.25.
[00245] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form providing a Cmax of abusable drug at 2 to about 10 hours; and said dosage form providing a therapeutic effect for at least about 12 hours. In other preferred embodiments, the dosage form provides a Cmax of abusable drug at about 2 to aboμt 8 hour or about 2 to about 6 hours, or about 2 to about 5 hours, or about 2 to about 7 hours, or about 2 to about 4.5 hours, or about 2 to about 4 hours, or 2 to about 3.5 hours, or about 2 to about 3 hours, or about 3 to about 10 hours, or about 3.5 to about 10 hours, or about 4 to about 10 hours, or about 4.5 to about 10 hours, or about 5 to about 10 hours, or 5 to about 10 hours, or about 6 to about 10 hours, or about 3 to about 8 hours, or about 3 to about 7 hours, or about 3 to about 6 hours, or about 4 to about 8 hours, or about 4 to about 6.
[00246] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form providing a Ci2/CmaX ratio of abusable drug of 0.1 to about 1; and said dosage form providing a therapeutic effect for at least about 12 hours. In other preferred embodiments, the dosage form provides a C)2/Cmax ratio of abusable drug of about 0.25 to about 0.9, or about 0.25 to about 0.8, or about 0.25 to about 0.75, or about 0.25 to about 0.6, or 0.25 to about 0.5, or about 0.25 to about 0.4, or about 0.25 to about 0.35, or about 0.3 to about 0.95, or about 0.4 to about 0.95, or about 0.5 to about 0.95, or about 0.65 to about 0.95, or about 0.75 to about 0.95, or about 0.3 to about 0.8, or about 0.4 to about 0.75, or about 0.5 to about 0.75, or about 0.1 to about 0.9, or about 0.1 to about 0.8, or about 0.1 to about 0.7, or about 0.1 to about 0.6, or about 0.1 to about 0.5, or about 0.1 to about 0.4, or about 0.1 to about 0.3, or about 0.2 to about 1.0, or about 0.25 to about 1.0, or about 0.4 to about 1.0, or about 0.5 to about 0.9, or about 0.5 to about 0.85, or about 0.5 to about 0.8, or about 0.5 to about 0.75, or about 0.5 to about 1.0, or about 0.65 to about 1.0, or about 0.75 to about 1.0, or about 0.2 to about 0.9, or about 0.3 to about 0.95, or about 0.3 to about O.85,or about 0.3 to about 0.8, or about 03 to about 0.75, or about 0.3 to about 0.7, or about 0.3 to about 0.6, or about 0.4 to about 0.9, or about 0.4 to about 0.8, or about 0.4 to about 0.7, or about 0.4 to about 0.6, or about 0.8 to about 1, or about 0.8 to about 1.1.
[00247] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form providing a percent fluctuation of abusable drug of less than 400%; and said dosage form providing a therapeutic effect for at least about 12 hours. In other preferred embodiments, the dosage form provides a percent fluctuation of abusable drug of less than about 375%, or less than about 350%, or less than about 325%, or less than about 300%, or less than about 275%, or less than about 250%, or less than about 225%, or less than about 200%, or less than about 175%, or less than about 150%, or less than about 125%, or less than about 100%, or less than about 75%; or less than about 50%, or less than about 25%.
[00248] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form after administration to a human patient, providing a W50 of abusable drug of 2 to about 11 hours; and said dosage form providing a therapeutic effect for at least about 12 hours. In other preferred embodiments, the dosage form provides a W50 of abusable drug of about 2 to about 10 hours, or about 2 to about 9 hours, or about 2 to about 9 hours, or about 2 to about 8 hours, or 2 to about 7 hours, or. about 2 to about 6 hours, or about 2 to about 5 hours, or about 2 to about 4 hours, or about 3 to about 10 hours, or about 4 to about 10 hours, or about 5 to about 10 hours, or about 6 to about 10 hours, or 7 to about 10 hours, or about 3 to about 8 hours, or about 4 to about 8 hours, or about 4 to about 7 hours, or about 3 to about 6 hours.
[00249] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form after administration to a human patient, providing a HVD of abusable drug of 1.5 to about 10 hours; and said dosage form providing a therapeutic effect for at least about 12 hours. In other preferred embodiments, the dosage form provides an HVD of abusable drug of about 1.5 to about 9 hours, or about 1.5 to 8 hours, or about 1.5 to about 7 hours, or about 1.5 to 6 hours, or about 1.5 to about 5 hours, or about 1.5 to about 4 hours, or about 2 to about 10 hours, or about 3 to 10 hours, or about 4 to about 10 hours, or about 5 to 10 hours, or about 6 to about 10 hours, or about 8 to 10 hours, about 3 to about 8 hours, or about 4 to 8 hours, or about 5 to about 7 hours, or about 3 to .6 hours, or about 3 to about 8 hours, or about 5 to about 8 hours.
[00250] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient, said dosage form after administration to a human patient, providing an AI of abusable drug of not more that 4.0; and said dosage form providing a therapeutic effect for at least about 12 hours. In other preferred embodiments, the dosage form provides an AI of abusable drug of not more than about 3.75, or not more than about 3.5, or not more than about 3.25, or not more than about 3, or not more than about 2.75, or not more than about 2.5, or not more than about 2, or not more than about 1.5, not more than about 1.25, or not more than about 1, or not more than about 0.75.
[00251] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours. In other preferred embodiments, the dosage form provides said an in-vitro release rate of from 0% to about 40% at 1 hour, from about 5% to about 55% at 2 hours, from about 10% to about 60% at 4 hours, from about 15% to about 70% at 6 hours, from about 25% to about 80% at 9 hours, and greater than about 50% at 12 hours.
[00252] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours; said dosage form providing a Cmax from a mean of about 2 to about 10 hours after first administration or at steady state.
[00253} In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours-, and greater than about 65% at 12 hours; said dosage form providing a Cmin occurring from a mean of about 10 to about 14 hours after first administration or at steady state. [00254] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours; said dosage form providing a mean abusable drug AUCoVAUCo- ratio after first administration of about 0.4, or about 0.5, or about 0.6, or about 0.7, or about 0.75, or about 0.8, or about 0.85, or about 0.88, or about 0.90, or about 0.92, or about 0.95, or about 0.97 or about 0.99.
[00255] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for twice-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours; said in-vitro release rate being substantially independent of pH in that a difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH, when measured in-vitro using the USP Basket and Paddle Methods of USP Drug Release test of U.S. Pharmacopeia (2003) at 100 rpm in 900 ml aqueous buffer, is no greater than 30%. [00256] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a Cmax of abusable drug at about 3 to about 20 hours; and said dosage form providing a therapeutic effect for at least about 24 hours. In some preferred embodiments, the abusable drugs dosage forms provide a Cmax of abusable drug at about 3 to about 18 hours, or about 3 to about 15 hours, or about 3 to about 12 hours, or at about 3 to about 10 hours, or at about 3 to about 8 hours, or at about 3 to about 7 hours, or at about 3 to about 7 hours, or about 4 to about 20 hours, or about 5 to about 20 hours, or about 6 to about 20 hours, or at about 8 to about 20 hours, or at about 10 to about 20 hours, or at about 12 to about 20 hours, or at about 14 to about 20 hours, or about 18 to about 20 hours, or about 4 to about 18 hours, or about 4 to about 16 hours, or at about 4 to about 12 hours, or at about 4 to about 8 hours, or at about 4 to about 10 hours, or at about 3 to about 6 hours.
[00257] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a Cmin of abusable drug at about 20 to about 28 hours; and said abusable drugs dosage forms providing a therapeutic effect for at least about 24 hours. In some preferred embodiments, the abusable drugs dosage forms provide a Cmin of abusable drug at about 20 to about 26 hours, or about 20 to about 27 hours, or about 20 to about 25 hours, or about 20 to about 24 hours, or about 20 to about 23 hours, or about 21 to about 28 hours, or about 22 to about 28 hours, or about 23 to about 28 hours, or about 23.5 to about 28 hours, or about 22 to 26 hours.
[00258] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage from providing a Cmaχ of abusable drug from about 0.25 hours to about 30 hours.
[00259] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER; said dosage from providing a Cmin of abusable drug from about 0.5 hour to about 30 hours.
[00260] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a C24ZCm3x ratio of abusable drug of 0.1 to about 1; and said dosage form providing a therapeutic effect for at least about 24 hours. In other preferred embodiments, the dosage form provides a C24/Cmax ratio of abusable drug of about 0.25 to about 0.9, or about 0.25 to about 0.8, or about 0.25 to about 0.75, or about 0.25 to about 0.6, or 0.25 to about 0.5, or about 0.25 to about 0.4, or about 0.25 to about 0.35, or about 0.3 to about 0.95, or about 0.4 to about 0.95, or about 0.5 to about 0.95, or about 0.65 to about 0.95, or about" 0.75 to about 0.95, or about 0.3 to about 0.8, or about 0.4 to about 0.75, or about 0.5 to about 0.75, or about 0.1 to about 0.9, or about 0.1 to about 0.8, or about 0.1 to about 0.7, or about 0.1 to about 0.6, or about 0.1 to about 0.5, or about 0.1 to about 0.4, or about 0.1 to about 0.3, or about 0.2 to about 1.0, or about 0.25 to about 1.0, or about 0.4 to about 1.0, or about 0.5 to about 0.9, or about 0.5 to about 0.85, or about 0.5 to about 0.8, or about 0.5 to about 0.75, or about 0.5 to about 1.0, or about 0.65 to about 1.0, or about 0.75 to about 1.0, or about 0.2 to about 0.9, or about 0.3 to about 0.95, or about 0.3 to about 0.85,or about 0.3 to about 0.8, or about 03 to about 0.75, or about 0.3 to about 0.7, or about 0.3 to about 0.6, or about 0.4 to about 0.9, or about 0.4 to about 0.8, or about 0.4 to about 0.7, or about 0.4 to about 0.6, or about 0.8 to about 1, or about 0.8 to about 1.1. [00261] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form providing a percent fluctuation of abusable drug of less than 400%; and said dosage form providing a therapeutic effect for at least about 24 hours. In other preferred embodiments, the dosage form provides a percent fluctuation of abusable drug of less than about 375%, or less than about 350%, or less than about 325%, or less than about 300%, or less than about 275%, or less than about 250%, or less than about 225%, or less than about 200%, or less than about 175%, or less than about 150%, or less than about 125%, or less than about 100%, or less than about 75%, or less than about 50%, or less than about 25%.
[00262] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said abusable drugs dosage form after administration to a human patient, providing a W50 of abusable drug of 4 to about 22 hours; and said dosage form providing a therapeutic effect for at least about 24 hours. In other preferred embodiments, the abusable drugs dosage from provides a W50 of abusable drug of about 4 to about 20 hours, or about 4 to about 19 hours, or about 4 to about 18 hours, or 4 to about 16 hours, or 4 to about 14 hours, or about 4 to about 12 hours, or about 4 to about 10 hours, or about 4 to about 8 hours, or about 6 to about 20 hours, or about 8 to about 20 hours, or about 10 to about 20 hours, or about 12 to about 20 hours, or 14 to about 20 hours, or about 6 to about 16 hours, or about 8 to about 16 hours, or about 8 to about 14 hours, or about 6 to about 12 hours.
[00263] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said abusable drugs dosage form after administration to a human patient, providing a HVD of abusable drug of 3 to about 20 hours; and said dosage form providing a therapeutic effect for at least about 24 hours. In other preferred embodiments, the abusable drugs dosage from provides an HVD of abusable drug of about 3 to about 18 hours, or about 3 to 16 hours, or about 3 to about 14 hours, or about 3 to 12 hours, or about 3 to about 10 hours, or about 3 to about 8 hours, or about 4 to about 20 hours, or about 6 to 20 hours, or about 8 to about 20 hours, or about 10 to 20 hours, or about 12 to about 20 hours, or about 16 to 20 hours, about 6 to about 16 hours, or about 8 to 16 hours, or about 10 to about 14 hours, or about 6 to 12 hours, or about 6 to about 16 hours, or about 10 to about 16 hours.
[00264] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient, said dosage form after administration to a human patient, providing an AI of abusable drug of not more that 4.0; and said abusable drugs dosage form providing a therapeutic effect for at least about 24 hours, hi other preferred embodiments, the abusable drugs dosage from provides an AI of abusable drug of not more than about 3.75, or not more than about 3.5, or not more than about 3.25, or not more than about 3, or not more than about 2.75, or not more than about 2.5, or not more than about 2, or not more than about 1.5, not more than about 1.25, or not more than about 1, or not more than about 0.75.
[00265] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said abusable drug dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 °C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours.
[00266] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said abusable drug dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said dosage form providing a Cm3x from a mean of about 3 to about 20 hours after first administration or at steady state.
[00267] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said dosage form providing a Cmin of abusable drug occurring from a mean of about 20 to about 28 hours after first administration or at steady state.
[00268] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said dosage form providing a mean abusable drug AUCo-t/AUCo-∞ ratio after first administration of about 0.4, or about 0.5, or about 0.6, or about 0.7, or about 0.75, or about 0.8, or about 0.85, or about 0.88, or about 0.90, or about 0.92, or about 0.95, or about 0.97 or about 0.99.
[00269] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for once-a-day administration to a human patient; said dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of from 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours; said in-vitro release rate being substantially independent of pH in that a difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH, when measured in-vitro using the USP Basket and Paddle Methods of USP Drug Release test of U.S. Pharmacopeia (2003) at 100 rpm in 900 ml aqueous buffer, is no greater than 30%.
[00270] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for extended release administration to a human patient; said dosage form after administration to a human patient providing a mean abusable drug Cmax occurring from a mean of about 0.25 to about 22 hours; said dosage form providing a mean abusable drug Cmjn occurring from a mean of about 0.5 to about 28 hours; said dosage form providing a mean abusable drug HVD of about 1 to about 5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug W50 of about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug AI of not more than 3.0; said dosage form providing a mean abusable drug percent fluctuation of less than 400%; said dosage form providing a mean abusable drug Cmin/Cmaχ ratio of 0.1 to about 1.0; said dosage form providing at least 80% of the steady state abusable drug therapeutic concentration after administration of < three doses at their intended dosing frequency; said dosage form providing a mean abusable drug AUCo-t to AUCo-~ ratio of greater than 0.4; and said dosage form providing a mean time to 75% abusable drug Cmax of about 100% to about 2000% of the time to 75% mean Cmax of an oral immediate release abusable drug solution or suspension.
[00271] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for extended release administration to a human patient; said dosage form after administration to a human patient providing a mean abusable drug Cm3x which is less than 65% of the Cmaχ of an equivalent dose of an oral immediate release abusable drug solution or suspension; and said dosage form maintaining a mean abusable drug plasma concentration within 50% of Cmaχ for about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action.
[00272] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER to render said dosage form abuse deterrent and/or suitable for extended release administration to a human patient; said dosage form after administration to a human patient providing a mean abusable drug Cm3x occurring from a mean of about 0.25 to about 22 hours; said dosage form providing a mean abusable drug Cmin occurring from a mean of about 0.5 to about 28 hours; said dosage form providing a mean abusable drug HVD of about 1 to about 5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug W50 of about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action; said dosage form providing a mean abusable drug AI of not more than 3.0; said dosage form providing a mean abusable drug percent fluctuation of less than 400%; said dosage form providing a mean abusable drug Cmin/Cmax ratio of 0.1 to about 1.0; said dosage form providing at least 80% of the steady state abusable drug therapeutic concentration after administration of < three doses at their intended dosing frequency; said dosage form providing a mean abusable drug AUC0.t to AUCo-∞ ratio of greater than 0.4; said dosage form providing a mean time to 75% abusable drug Cmax of about 100% to about 2000% of the time to 75% mean Cmax of an oral immediate release abusable drug solution or suspension; said dosage form after administration to a human patient providing a mean abusable drug Cmax which is less than 65% of the Cmax of an equivalent dose of an oral immediate release abusable drug solution or suspension; and said dosage form maintaining a mean abusable drug plasma concentration within 50% of Cmax for about 1 to about 5.5 hours for each 6 hour time period of intended dosing frequency and intended duration of action.
[00273] In some preferred embodiments, the abusable drugs dosage forms provide an in- vitro release of from 0% to about 50% by weight of the abusable drug or a pharmaceutically acceptable salt thereof from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) at 37 0C. In other preferred embodiments, said in-vitro release rate by weight of the abusable drug or a pharmaceutically acceptable salt thereof from said dosage form is from about 5% to about 45% , or about 10% to about 50%, or about 5% to about 60%, or about 5% to about 70%, or about 5% to about 80%, or about 5% to about 90%, or about 5% to about 100%, or about 10% to about 20%, or about 10% to about 35% , or about 10% to about 50%, or about 10% to about 60%, or about 10% to about 70%, or about 10% to about 80%, or about 10% to about 90%, or about 10% to about 100%, or about 20% to about 40% , or about 20% to about 50%, or about 20% to about 60%, or about 20% to about 70%, or about 20% to about 80%, or about 20% to about 90%, or about 20% to about 100%, or about 30% to about 50%, or about 30% to about 60%, or about 30% to about 70%, or about 30% to about 80%, or about 30% to about 90%, or about 40% to about 80%, or about 40% to about 90%, or about 60% to about 100%, or greater than about 5%, or greater than about 10%, or greater than about 15%, or greater than about 20%, or greater than about 30%, or greater than about 40%, or greater than about 50%, or greater than about 60%, or greater than about 80%, or greater than about 90%, or greater than about 95%, at one hour, when measured by the USP Basket and Paddle Methods at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) at 37 0C.
[00274] In some preferred embodiments, the abusable drugs dosage form provides a CmaX of abusable drug which is less than 65% of the Cmax of an equivalent dose of an oral immediate release abusable drug solution or suspension. In other preferred embodiments, said dosage form provides a Cmax which is less than about 85%, or less than about 75%, or less than about 60%, or less than about 55%, or less than about 50%, or less than about 45%, or less than about 40%, or less than about 30%, or less than about 20% of the Cmax of an equivalent dose of an oral immediate release abusable drug solution or suspension.
[00275] In some preferred embodiments, the dosage form provides a time to
75% mean Cm3x of abusable drug which is about 100% to about 2000% of the time to 75% mean Cmax of an oral immediate release abusable drug solution or suspension. [00276] In some preferred embodiments, the dosage form provides a time to
30% mean Cmax of abusable drug which is about 100% to about 2000% of the time to 30% mean Cmax of an oral immediate release abusable drug solution or suspension.
[00277] In some preferred embodiments, the dosage from maintains a plasma abusable drug concentration within 50% of Cmax for about 1 to about 9 hours during a 12 hour dosing interval. In other preferred embodiments, said dosage form maintains plasma abusable drug concentration within 50% of Cmax for about 2 to about 9 hours, or about 3 to about 9 hours, or about 4 to about 9 hours, or about 5 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 11 hours, or about 2 to about 11 hours, or about 3 to about 11 hours or about 4 to about 11 hours, or about 5 to about 11 hours, or about 6 to about 11 hours, or about 7 to about 11 hours, or about 8 to about 11 hours, or about 1 to about 10 hours, or about 2 to about 10 hours, or about 3 to about 10 hours or about 4 to about 10 hours, or about 5 to about 10 hours, or about 6 to about 10 hours, or about 7 to about 10 hours, or about 8 to about 10 hours, or about 1 to about 7 hours, or about 2 to about 7 hours, or about 3 to about 7 hours or about 4 to about 7 hours, or about 5 to about 7 hours, or about 6 to about 7 hours, or about 1 to about 4 hours, or about 1 to about 5 hours, during a 12 hour dosing interval.
[00278] In some preferred embodiments, the dosage from maintains a plasma abusable drug concentration within 30% of Cmax for about 1.5 to about 9 hours during a 12 hour dosing interval. In other preferred embodiments, said dosage form maintains plasma abusable drug concentration within 30% of Cmax for about 2 to about 9 hours, or about 3 to about 9 hours, or about 4 to about 9 hours, or about 5 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 11 hours, or about 2 to about 11 hours, or about 3 to about 11 hours or about 4 to about 11 hours, or about 5 to about 11 hours, or about 6 to about 11 hours, or about 7 to about 11 hours, or about 8 to about 11 hours, or about 1 to about 10 hours, or about 2 to about 10 hours, or about 3 to about 10 hours or about 4 to about 10 hours, or about 5 to about 10 hours, or about 6 to about 10 hours, or about 7 to about 10 hours, or about 8 to about 10 hours, or about 1 to about 7 hours, or about 2 to about 7 hours, or about 3 to about 7 hours or about 4 to about 7 hours, or about 5 to about 7 hours, or about 6 to about 7 hours, or about 1 to about 4 hours, or about 1 to about 5 hours, during a 12 hour dosing interval.
[00279] In some preferred embodiments, the dosage from maintains a plasma abusable drug concentration within 65% of Cmaχ for about 1 to about.9 hours during a 12 hour dosing interval. In other preferred embodiments, said dosage form maintains plasma abusable drug concentration within 65% of Cmax for about 2 to about 9 hours, or about 3 to about 9 hours, or about 4 to about 9 hours, or about 5 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 11 hours, or about 2 to about 11 hours; or about 3 to about 11 hours or about 4 to about 11 hours, or about 5 to about 11 hours, or about 6 to about 11 hours, or about 7 to about 11 hours, or about 8 to about 11 hours, or about 1 to about 10 hours, or about 2 to about 10 hours, or about 3 to about 10 hours or about 4 to about 10 hours, or about 5 to about 10 hours, or about 6 to about 10 hours, or about 7 to about 10 hours, or about 8 to about 10 hours, or about 1 to about 7 hours, or about 2 to about 7 hours, or about 3 to about 7 hours or about 4 to about 7 hours, or about 5 to about 7 hours, or about 6 to about 7 hours, or about 1 to about 4 hours, or about 1 to about 5 hours, during a 12 hour dosing interval.
[00280] In some preferred embodiments, the dosage from maintains a plasma abusable drug concentration within 55% of Cmax for about 3 to about 22 hours during a 24 hour dosing interval. In other preferred embodiments, said dosage form maintains plasma abusable drug concentration within 50% of Cm3x for about 1 to about 9 hours, or about 4 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 20 hours, or about 2 to about 20 hours, or about 3 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours or about 2 to about 18 hours, or about 2 to about 16 hours, or about 1 to about 14 hours, or about 1 to about 12 hours, or about 4 to about 16 hours, or about 4 to about 18 hours, or about 4 to about 20 hours, or about 3 to about 15 hours or about 6 to about 15 hours, or about 6 to about 12 hours, or about 6 to about 18 hours, or about 6 to about 20 hours, or about 5 to about 12 hours, or about 5 to about 14 hours, or about 3 to about 22 hours, or about 3 to about 9 hours or about 3 to about 12 hours, or about 1 to about 6 hours, or about 2 to about 8 hours, or about 2 to about 10 hours, or about 3 to about 16 hours, during a 24 hour dosing interval.
[00281] In some preferred embodiments, the dosage from maintains a plasma abusable drug concentration within 30% of Cmaχ for about 2 to about 22 hours during a 24 hour dosing interval. In other preferred embodiments, said dosage form maintains plasma abusable drug concentration within 30% of Cn^x for about 1 to about 9 hours, or about 4 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 20 hours, or about 2 to about 20 hours, or about 3 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours or about 2 to about 18 hours, or about 2 to about 16 hours, or about 1 to about.14 hours, or about 1 to about 12 hours, or about 4 to about 16 hours, or about 4 to about 18 hours, or about 4 to about 20 hours, or about 3 to about 15 hours or about 6 to about 15 hours, or about 6 to about 12 hours, or about 6 to about 18 hours, or about 6 to about 20 hours, or about 5 to about 12 hours, or about 5 to about 14 hours, or about 3 to about 22 hours, or about 3 to about 9 hours or about 3 to about 12 hours, or about 1 to about 6 hours, or about 2 to about 8 hours, or about 2 to about 10 hours, or about 3 to about 16 hours, during a 24 hour dosing interval.
[00282] In some preferred embodiments, the dosage from maintains a plasma abusable drug concentration within 65% of Cmaχ for about 2 to about 22 hours during' a 24 hour dosing interval. In other preferred embodiments, said dosage form maintains plasma abusable drug concentration within 65% of Cmax for about 1 to about 9 hours, or about 4 to about 9 hours, or about 6 to about 9 hours, or about 1 to about 20 hours, or about 2 to about 20 hours, or about 3 to about 20 hours, or about 1 to about 18 hours, or about 1 to about 16 hours or about 2 to about 18 hours, or about 2 to about 16 hours, or about 1 to about 14 hours, or about 1 to about 12 hours, or about 4 to about 16 hours, or about 4 to about 18 hours, or about 4 to about 20 hours, or about 3 to about 15 hours or about 6 to about 15 hours, or about 6 to about 12 hours, or about 6 to about 18 hours, or about 6 to about 20 hours, or about 5 to about 12 hours, or about 5 to about 14 hours, or about 3 to about 22 hours, or about 3 to about 9 hours or about 3 to about 12. hours, or about 1 to about 6 hours, or about 2 to about 8 hours, or about 2 to about 10 hours, or about 3 to about 16 hours, during a 24 hour dosing interval.
[00283] In some preferred embodiments, the dosage form provides a Tmax of abusable drug at a time point 1 to 18 times later than the TmaX provided by an equivalent dose of an oral immediate release abusable drug solution or suspension, hi the dosage form provides a Tmax at a time point about 1 to 15 times late, or about of 1 to 10 times later, or about of 1 to 7 times later, or about of 1 to 4 times later, or about of 3 to 20 times later, or about of 3 to 10 times later, or about of 3 to 5 times later, or about 1.5 to 15 times later, or about of 1.5 to 10 times later, or about of 1.5 to 7 times later, or about of 1.5 to 3 times later, or about of 2 to 20 times later, or about of 2 to 10 times later, or about of 2 to 5 times later, or about of 2 to 3 times later, or about of 2.5 to 20 times later, or about of 2.5 to 8 times later, or about of 2.5 to 5 times later, or about of 2.5 to 4 times later, or about of 3 to 20 times later, or about of 3 to 10 times later, or about of 3 to 5 times later.
[00284] In some preferred embodiments, the dosage form provides a mean in vivo extent of absorption of abusable drug from 0 to 4 hours which is at least 20% of the mean in vivo extent of absorption from to 0 to 12 hours, wherein the mean in vivo extent of absorption is the area under the plasma or serum abusable drug concentration time curve from the time of drug administration to the specified time point. In other preferred embodiments, said in vivo extent of absorption from 0 to 4 hours is at least about 5%, or at least about 10%, or at least about 15%, or at least about 25%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, at least about 90%, or about 100% of the mean in vivo extent of absorption from to 0 to 12 hours.
[00285] In some preferred embodiments, the dosage form provides a mean in vivo extent of absorption of abusable drug from 0 to 8 hours which is at least 20% of the mean in vivo extent of absorption from to 0 to 24 hours, wherein the mean in vivo extent of absorption is the area under the plasma or serum abusable drug concentration time curve from the time of drug administration to the specified time point. In other preferred embodiments, said in vivo extent of absorption from 0 to 8 hours is at least about 5%, or at least about 10%, or at least about 15%, or at least about 25%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, at least about 90%, or about 100% of the mean in vivo extent of absorption from to 0 to 24 hours.
[00286] In some preferred embodiments, the dosage form provides a mean in vivo extent of absorption of abusable drug from 0 to 12 hours which is at least 20% of the mean in vivo extent of absorption from to 0 to 24 hours, wherein the mean in vivo extent of absorption is the area under the plasma or serum abusable drug concentration time curve from the time of drug administration to the specified time point, hi other preferred embodiments, said in vivo extent of absorption from 0 to 12 hours is at least about 5%, or at least about 10%, or at least about 15%, or at least about 25%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, at least about 90%, or about 100% of the mean in vivo extent of absorption from to 0 to 24 hours.
[00287] In some preferred embodiments, the dosage form provides a mean in vivo extent of absorption of abusable drug over the dosing interval, AUCo-t (e.g., from 0 to 8 hours, or from 0 to 12 hours or from 0 to 24 hours) which is at least 40% of the mean in vivo extent of absorption from to 0 to infinity (AUCo-). In other preferred embodiments, said AUCo-t is at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% of the mean in vivo extent of absorption from to 0 to infinity
[00288] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% to about 100% at 0.5 hours, and greater than about 60% at 1 hour.
[00289] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% to about 40% at 1 hour, from about 5% to about 60% at 2 hours, from about 10% to about 75% at 4 hours, from about 20% to about 75% at 6 hours, from about 30% to about 80% at 9 hours, and greater than about 70% at 12 hours.
[00290] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 1% and about 45% at 1 hour, between about 5% and about 70% at 2 hours, between about 10% and about 90% at 4 hours, between about 20% and about 90% at 8 hours, greater than about 60% at 12 hours, greater than about 80% at 18 hours, and greater than about 85% at 24 hours.
[00291] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between(1.6 and 7.2 at 37 0C of between 5% and about 60% at 1 hour, between about 12.5% and about 80% at 2 hours, between about 25% and about 95% at 4 hours, between about 45% and about 100% at 8 hours, greater than about 55% at 12 hours, greater than about 65% at 18 hours, and greater than about 70% at 24 hours. [00292] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 °C of between 0% and about 40% at 1 hour, between about 0% and about 70% at 2 hours, between about 5% and about 95% at 4 hours, between about 12.5% and about 100% at 8 hours, between about 20% and about 100% at 12 hours, between about 35% and about 100% at 16 hours, between about 55% and about 100% at 24 hours, and greater than about 75% at 36 hours.
[00293] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% and about 60% at 1 hour, between about 0% and about 75% at 2 hours, between about 5% and about 95% at 4 hours, between about 12.5% and about 100% at 8 hours, between about 15% and about 100% at 12 hours, between about 25% to about 100% at 16 hours, between about 30% and about 100% hours at 24 hours and greater than 60% at 36 hours.
[00294] hi some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) at 37 0C of between 0% to about 50% by weight of the abusable drug. In other preferred embodiments, said release rate is between 0% to about 1%, or 0% to about 3%, or 0% to about 5%, or 0% to about 10%, or 0% to about 15%, or 0% to about 20%, 0% to about 30%, or 0% to about 40%, or 0% to about 60%, or 0% to about 70%, or 0% to about 80%, or 0% to about 90%, 0% to about 100%.
[00295] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% and about 60% at 1 hour, between about 0% and about 80% at 2 hours, between about 3% and about 95% at 4 hours and between about 10% and about 100% at 8 hours. In other preferred embodiments, said release rate is between 0% and about 10% at 1 hour, between about 0% and about 20% at 2 hours, between about 2% and about 80% at 4 hours and between about 5% and about 100% at 8 hours; or between 0% and about 20% at 1 hour, between about 0% and about 40% at 2 hours, between about 0% and about 80% at 4 hours and between about 2% and about 100% at 8 hours; or between 0% and about 40% at 1 hour, between about 0% and about 60% at 2 hours, between about 5% and about 85% at 4 hours and between about 5% and about 90% at 8 hours and greater than 20% at 12 hours; or between 0% and about 50% at 1 hour, between about 0% and about 50% at 2 hours, between about 10% and about 90% at 4 hours and between about 15% and about 90% at 8 hours and greater than 30% at 12 hours; -or between 0% and about 70% at 1 hour, between about 0% and about 70% at 2 hours, between about 10% and about 75% at 4 hours and between about 15% and about 90% at 8 hours and greater than 30% at 12 hours.
[00296] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 10% and about 65% at 1 hour, between about 20% and about 75% at 2 hours, between about 30% and about 95% at 4 hours and between about 40% and about 100% at 8 hours. In other preferred embodiments, said release rate is between 2% and about 70% at 1 hour, between about 5% and about 80% at 2 hours, between about 10% and about 90% at 4 hours and between about 20% and about 100% at 8 hours; or between 5% and about 60% at 1 hour, between about 10% and about 75% at 2 hours, between about 15% and about 85% at 4 hours and between about 30% and about 100% at 8 hours; or between 20% and about 70% at 1 hour, between about 20% and about 75% at 2 hours, between about 20% and about 90% at 4 hours and between about 40% and about 100% at 8 hours; or between 30% and about 80% at 1 hour, between about 40% and about 85% at 2 hours, between about 40% and about 90% at 4 hours and between about 60% and about 100% at 8 hours; or between 1% and about 20% at 1 hour, between about 5% and about 20% at 2 hours, between about 10% and about 40% at 4 hours and between about 20% and about 40% at 8 hours and greater than 40% at 12 hours. ] hi some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25%' to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours. In other preferred embodiments, said release rate is between 0% to about 30% at 1 hour, from about 5% to about 45% at 2 hours, from about 10% to about 60% at 4 hours, from about 15% to about 70% at 6 hours, from about 25% to about 80% at 9 hours, and greater than about 50% at 12 hours; or between 0% to about 20% at 1 hour, from about 2% to about 35% at 2 hours, from about 5% to about 50% at 4 hours, from about 10% to about 60% at 6 hours, from about 15% to about 70% at 9 hours, and greater than about 40% at 12 hours; or between 0% to about 10% at 1 hour, from about 1% to about 30% at 2 hours, from about 5% to about 40% at 4 hours, from about 10% to about 60% at 6 hours, from about 15% to about 70% at 9 hours, and greater than about 40% at 12 hours; or between 0% to about 5% at 1 hour, from about 0% to about 10% at 2 hours, from about 2% to about 20% at 4 hours, from about 5% to about 30% at 6 hours, from about 10% to about 40% at 9 hours, and greater than about 30% at 12 hours; or between 0% to about 50% at 1 hour, from about 15% to about 70% at 2 hours, from about 20% to about 75% at 4 hours, from about 30% to about 80% at 6 hours, from about 30% to about 90% at 9 hours, and greater than about 70% at 12 hours; or between 0% to about 60% at 1 hour, from about 15% to about 80% at 2 hours, from about 25% to about 85% at 4 hours, from about 35% to about 90% at 6 hours, from about 40% to about 90% at 9 hours, and greater than about 80% at 12 hours; ; or between 0% to about 70% at 1 hour, from about 20% to about 80% at 2 hours, from about 25% to about 80% at 4 hours, from about 35% to about 80% at 6 hours, from about 40% to about 80% at 9 hours, and greater than about 60% at 12 hours; or between 0% to about 75% at 1 hour, from about 30% to about 80% at 2 hours, from about 35% to about 90% at 4 hours, from about 50% to about 90% at 6 hours, from about 55% to about 95% at 9 hours, and greater than about 70% at 12 hours. ] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between L6 and 7.2 at 37 0C of between 5% and about 50% at 1 hour, between about 10% and about 75% at 2 hours, between about 20% and about 95% at 4 hours, between about 40% and about 100% at 8 hours, greater than about 50% at 12 hours, greater than about 70% at 18 hours, and greater than about 80% at 24 hours. In other preferred embodiments, said release rate is between 2% and about 50% at 1 hour, between about 5% and about 75% at 2 hours, between about 15% and about 75% at 4 hours, between about 30% and about 90% at 8 hours, greater than about 40% at 12 hours, greater than about 60% at 18 hours, and greater than about 70% at 24 hours; or between 1% and about 40% at 1 hour, between about 2% and about 60% at 2 hours, between about 10% and about 65% at 4 hours, between about 20% and about 80% at 8 hours, greater than about 30% at 12 hours, greater than about 40% at 18 hours, and greater than about 60% at 24 hours; or between 5% and about 60% at 1 hour, between about 15% and about 80% at 2 hours, between about 25% and about 95% at 4 hours, between about 45% and about 100% at 8 hours, greater than about 60% at 12 hours, greater than about 80% at 18 hours, and greater than about 90% at 24 hours; or between 10% and about 65% at 1 hour, between about 20% and about 85% at 2 hours, between about 30% and about 100% at 4 hours, between about 60% and about 100% at 8 hours, greater than about 70% at 12 hours, greater than about 90% at 18 hours, and greater than about 95% at 24 hours. ] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours. In other preferred embodiments, said release rate is between 0% to about 20% at 1 hour, from about 5% to about 50% at 4 hours, from about 10% to about 60% at 8 hours, from about 15% to about 70% at 12 hours, from about 25% to about 90% at 18 hours, and greater than about 55% at 24 hours; or between 0% to about 10% at 1 hour, from about 5% to about 40% at 4 hours, from about 8% to about 50% at 8 hours, from about 10% to about 60% at 12 hours, from about 22% to about 80% at 18 hours, and greater than about 45% at 24 hours; or between 0% to about 35% at 1 hour, from about 15% to about 70% at 4 hours, from about 25% to about 75% at 8 hours, . from about 30% to about 85% at 12 hours, from about 40% to about 100% at 18 hours, and greater than about 75% at 24 hours; or between 0% to about 40% at 1 hour, from about 20% to about 70% at 4 hours, from about 30% to about 80% at 8 hours, from about 35% to about 90% at 12 hours, from about 45% to about 100% at 18 hours, and greater than about 80% at 24 hours; or between 0% to about 45% at 1 hour, from about 25% to about 75% at 4 hours, from about 35% to about 85% at 8 hours, from about 40% to about 90% at 12 hours, from about 50% to about 100% at 18 hours, and greater than about 90% at 24 hours; or between 0% to about 50% at 1 hour, from about 30% to about 80% at 4 hours, from about 40% to about 90% at 8 hours, from about 45% to about 95% at 12 hours, from about 60% to about 100% at 18 hours, and greater than about 95% at 24 hours; or between 0% to about 60% at 1 hour, from about 40% to about 80% at 4 hours, from about 45% to about 90% at 8 hours, from about 50% to about 100% at 12 hours, from about 70% to about 100% at 18 hours, and greater than about 80% at 24 hours.
[00300] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 3% and about 95% at 4 hours, between about 10% and about 100% at 8 hours, between about 25% and about 100% at 12 hours, between about 30% and about 100% at 16 hours, between about 50% and about 100% at 24 hours, and greater than about 80% at 36 hours. In other preferred embodiments, said release rate is between 0% and about 40% at 1 hour, between about 0% and about 65% at 2 hours, between about 2% and about 85% at 4 hours, between about 8% and about 90% at 8 hours, between about 20% and about 95% at 12 hours, between about 25% and about 95% at 16 hours, between about 40% and about 90% at 24 hours, and greater than about 70% at 36 hours; or between 0% and about 30% at 1 hour, between about 0% and about 50% at 2 hours, between about 1% and about 75% at 4 hours, between about 5% and about 80% at 8 hours, between about 10% and about 85% at 12 hours, between about 15% and about 90% at 16 hours, between about 30% and about 80% at 24 hours, and greater than about 70% at 36 hours; or between 0% and about 60% at 1 hour, between about 0% and about 80% at 2 hours, between about 5% and about 100% at 4 hours, between about 15% and about 100% at 8 hours, between about 35% and about 100% at 12 hours, between about 40% and about 100% at 16 hours, between about 60% and about 100% at 24 hours, and greater than about 85% at 36 hours; or between 0% and about 65% at 1 hour, between about 0% and about 85% at 2 hours, between about 10% and about 100% at 4 hours, between about 20% and about 100% at 8 hours, between about 40% and about 100% at 12 hours, between about 50% and about 100% at 16 hours, between about 70% and about 100% at 24 hours, and greater than about 90% at 36 hours; or between 0% and about 70% at 1 hour, between about 0% and about 90% at 2 hours, between about 20% and about 100% at 4 hours, between about 30% and about 100% at 8 hours, between about 50% and about 100% at 12 hours, between about 60% and about 100% at 16 hours, between about 80% and about 100% at 24 hours, and greater than about 95% at 36 hours. ] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 20% and about 50% at 1 hour, between about 40% and about 75% at 2 hours, between about 60% and about 95% at 4 hours, between about 80% and about 100% at 8 hours and between about 90% and about 100% at 12 hours. In other preferred embodiments, said release rate is between 15% and about 45% at I hour, between about 35% and about 70% at 2 hours, between about 55% and about 90% at 4 hours, between about 75% and about 90% at 8 hours and between about 80% and about 95% at 12 hours; or between 10% and about 40% at 1 hour, between about 30% and about 65% at 2 hours, between about 50% and about 85% at 4 hours, between about 70% and about 85% at 8 hours and between about 75% and about 90% at 12 hours; or between 5% and about 35% at 1 hour, between about 25% and about 60% at 2 hours, between about 45% and about 80% at 4 hours, between about 65% and about 80% at 8 hours and between about 70% and about 85% at 12 hours; or between 25% and about 55% at 1 hour, between about 45% and about 80% at 2 hours, between about 65% and about 95% at 4 hours, between about 85% and about 100% at 8 hours and between about 95% and about 100% at 12 hours; or between 30% and about 60% at 1 hour, between about 50% and about 80% at 2 hours, between about 70% and about 95% at 4 hours, between about 90% and about 100% at 8 hours and between about 95% and about 100% at 12 hours; or between 35% and about 60% at 1 hour, between about 50% and about 80% at 2 hours, between about 80% and about 95% at 4 hours, between about 90% and about 100% at 8 hours and between about 95% and about 100% at 12 hours; or between 20% and about 40% at 1 hour, between about 40% and about 65% at 2 hours, between about 60% and about 85% at 4 hours, between about 70% and about 90% at 8 hours and between about 80% and about 100% at 12 hours. ] hi some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 10% and about 95% at 4 hours, between about 35% and about 100% at 8 hours, between about 55% and about 100% at 12 hours, between about 70% to about 100% at 16 hours, and greater than about 90% at 24 hours. In other preferred embodiments, said release rate is between 0% and about 40% at 1 hour, between about 0% and about 65% at 2 hours, between about 8% and about 85% at 4 hours, between about 30% and about 90% at 8 hours, between about 45% and about 100% at 12 hours, between about 60% to about 100% at 16 hours, and greater than about 80% at 24 hours; or between 0% and about 30% at 1 hour, between about 0% and about 55% at 2 hours, between about 5% and about 75% at 4 hours, between about 20% and about 80% at 8 hours, between about 35% and about 100% at 12 hours, between about 50% to about 100% at 16 hours, and greater than about 70% at 24 hours; or between 0% and about 20% at 1 hour, between about 0% and about 45% at 2 hours, between about 5% and about 65% at 4 hours, between about 10% and about 70% at 8 hours, between about 25% and about 80% at 12 hours, between about 40% to about 100% at 16 hours, and greater than about 60% at 24 hours; or between 0% and about 60% at 1 hour, between about 0% and about 80% at 2 hours, between about 15% and about 95% at 4 hours, between about 40% and about 100% at 8 hours, between about 60% and about 100% at 12 hours, between about 75% to about 100% at 16 hours, and greater than about 90% at 24 hours; or between 0% and about 65% at 1 hour, between about 0% and about 85% at 2 hours, between about 20% and about 90% at 4 hours, between about 45% and about 100% at 8 hours, between about 65% and about 100% at 12 hours, between about 80% to about 100% at 16 hours, and greater than about 90% at 24 hours; or between 0% and about 40% at 1 hour, between about 0% and about 50% at 2 hours, between about 10% and about 80% at 4 hours, between about 25% and about 70% at 8 hours, between about 40% and about 80% at 12 hours, between about 60% to about 100% at 16 hours, and greater than about 90% at 24 hours.] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at apH of between 1.6 and 7.2 at 37 0C of between 0% and about 30% at 1 hour, between about 0% and about 45% at 2 hours, between about 3% and about 55% at 4 hours, between about 10% and about 65% at 8 hours, between about 20% and about 75% at 12 hours, between about 30% to about 88% at 16 hours, between about 50% and about 100% hours at 24 hours and greater than 80% at 36 hours. In other preferred embodiments, said release rate is between 0% and about 25% at 1 hour, between about 0% and about 40% at 2 hours, between about 2% and about 50% at 4 hours, between about 8% and about 60% at 8 hours, between about 10% and about 70% at 12 hours, between about 25% to about 80% at 16 hours, between about 45% and about 100% hours at 24 hours and greater than 75% at 36 hours; or between 0% and about 20% at 1 hour, between about 0% and about 35% at 2 hours, between about 1% and about 45% at 4 hours, between about 5% and about 55% at 8 hours, between about 8% and about 65% at 12 hours, between about 20% to about 75% at 16 hours, between about 40% and about 100% hours at 24 hours and greater than 70% at 36 hours; or between 0% and about 15% at 1 hour, between about 0% and about 30% at 2 hours, between about 0% and about 40% at 4 hours, between about 5% and about 50% at 8 hours, between about 8% and about 60% at 12 hours, between about 15% to about 70% at 16 hours, between about 35% and about 100% hours at 24 hours and greater than 60% at 36 hours; or between 0% and about 10% at 1 hour, between about 0% and about 25% at 2 hours, between about 0% and about 35% at 4 hours, between about 5% and about 45% at 8 hours, between about 10% and about 50% at 12 hours, between about 10% to about 60% at 16 hours, between about 30% and about 90% hours at 24 hours and greater than 70% at 36 hours; or between 0% and about 35% at 1 hour, between about 0% and about 50% at 2 hours, between about 5% and about 60% at 4 hours, between about 15% and about 70% at 8 hours, between about 25% and about 80% at 12 hours, between about 35% to about 90% at 16 hours, between about 55% and about 100% hours at 24 hours and greater than 85% at 36 hours; or between 0% and about 40% at 1 hour, between about 0% and about 55% at 2 hours, between about 10% and about 65% at 4 hours, between about 20% and about 75% at 8 hours, between about 30% and about 85% at 12 hours, between about 40% to about 100% at 16 hours, between about 55% and about 100% hours at 24 hours and greater than 90% at 36 hours; or between 0% and about 45% at 1 hour, between about 0% and about 60% at 2 hours, between about 15% and about 70% at 4 hours, between about 25% and about 80% at 8 hours, between about 35% and about 90% at 12 hours, between about 45% to about 100% at 16 hours, between about 60% and about 100% hours at 24 hours and greater than 60% at 36 hours; or between 0% and about 50% at 1 hour, between about 5% and about 65% at 2 hours, between about 20% and about 75% at 4 hours, between about 30% and about 85% at 8 hours, between about 40% and about 95% at 12 hours, between about 50% to about 100% at 16 hours, between about 70% and about 100% hours at 24 hours and greater than 70% at 36 hours; or between 0% and about 30% at 1 hour, between about 5% and about 40% at 2 hours, between about 10% and about 60% at 4 hours, between about 20% and about 70% at 8 hours, between about 30% and about 100% at 12 hours, between about 40% to about 100% at 16 hours, between about 60% and about 100% hours at 24 hours and greater than 90% at 36 hours; or between 0% and about 30% at 1 hour, between about 0% and about 30% at 2 hours, between about 0% and about 30% at 4 hours, between about 5% and about 70% at 8 hours, between about 10% and about 80% at 12 hours, between about 20% to about 100% at 16 hours, between about 40% and about 100% hours at 24 hours and greater than 50% at 36 hours; or between 0% and about 20% at 1 hour, between about 0% and about 20% at 2 hours, between about 0% and about 20% at 4 hours, between about 0% and about 20% at 8 hours, between about 5% and about 40% at 12 hours, between about 10% to about 80% at 16 hours, between about 40% and about 100% hours at 24 hours and greater than 60% at 36 hours; or between 0% and about 10% at 1 hour, between about 0% and about 20% at 2 hours, between about 0% and about 40% at 4 hours, between about 5% and about 60% at 8 hours, between about 10% and about 80% at 12 hours, between about 20% to about 100% at 16 hours, between about 40% and about 100% hours at 24 hours and greater than 50% at 36 hours. ] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 3% and about 95% at 4 hours, between about 10% and about 100% at 8 hours, between about 20% and about 100% at 12 hours, between about 30% to about 100% at 16 hours, between about 50% and about 100% hours at 24 hours and greater than 80% at 36 hours, hi other preferred embodiments, said release rate is between 0% and about 45% at 1 hour, between about 0% and about 70% at 2 hours, between about 3% and about 90% at 4 hours, between about 8% and about 100% at 8 hours, between about 15% and about 100% at 12 hours, between about 25% to about 100% at 16 hours, between about 45% and about 100% hours at 24 hours and greater than 80% at 36 hours; or between 0% and about 40% at 1 hour, between about 0% and about 65% at 2 hours, between about 0% and about 80% at 4 hours, between about 5% and about 80% at 8 hours, between about 10% and about 90% at 12 hours, between about 20% to about 100% at 16 hours, between about 40% and about 100% hours at 24 hours and greater than 70% at 36 hours; or between 0% and about 35% at 1 hour, between about 0% and about 60% at 2 hours, between about 0% and about 70% at 4 hours, between about 3% and about 70% at 8 hours, between about 5% and about 80% at 12 hours, between about 15% to about 100% at 16 hours, between about 30% and about 100% hours at 24 hours and greater than 40% at 36 hours; or between 0% and about 60% at 1 hour, between about 0% and about 80% at 2 hours, between about 5% and about 100% at 4 hours, between about 15% and about 100% at 8 hours, between about 30% and about 100% at 12 hours, between about 40% to about 100% at 16 hours, between about 60% and about 100% hours at 24 hours and greater than 70% at 36 hours; or between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 5% and about 95% at 4 hours, between about 25% and about 80% at 8 hours, between about 30% and about 100% at 12 hours, between about 40% to about 100% at 16 hours, between about 60% and about 100% hours at 24 hours and greater than 60% at 36 hours; or between 0% and about 60% at 1 hour, between about 0% and about 85% at 2 hours, between about 5% and about 100% at 4 hours, between about 10% and about 100% at 8 hours, between about 20% and about 100% at 12 hours, between about 30% to about 100% at 16 hours, between about 50% and about 100% hours at 24 hours and greater than 80% at 36 hours. In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said abusable drugs dosage form providing an in-vitro release rate by weight of the abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and' 7.2 at 37 °C of between 15% and about 25% at 1 hour, between about 25% and about 35% at 2 hours, between about 30% and about 45% at 4 hours, between about 40% and about 60% at 8 hours, between about 55% and about 70% at 12 hours and between about 60% to about 75% at 16 hours. In other preferred embodiments, said release rate is between 10% and about 20% at 1 hour, between about 20% and about 30% at 2 hours, between about 25% and about 40% at 4 hours, between about 30% and about 50% at 8 hours, between about 50% and about 65% at 12 hours and between about 55% to about 65% at 16 hours; or between 5% and about 15% at 1 hour, between about 15% and about 25% at 2 hours, between about 20% and about 35% at 4 hours, between about 25% and about 45% at 8 hours, between about 45% and about 60% at 12 hours and between about 50% to about 60% at 16 hours; or between 15% and about 30% at 1 hour, between about 20% and about 40% at 2 hours, between about 20% and about 50% at 4 hours, between about 30% and about 70% at 8 hours, between about 60% and about 80% at 12 hours and between about 70% to about 90% at 16 hours; or between 0% and about 50% at 1 hour, between about 5% and about 50% at 2 hours, between about 5% and about 70% at 4 hours, between about 10% and about 80% at 8 hours, between about 20% and about 100% at 12 hours and between about 40% to about 100% at 16 hours; or between 15% and about 40% at 1 hour, between about 15% and about 45% at 2 hours, between about 20% and about 60% at 4 hours, between about 20% and about 80% at 8 hours, between about 30% and about 90% at 12 hours and between about 40% to about 100% at 16 hours.
[00306] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said in-vitro release rate being substantially independent of pH in that a difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH, when measured in-vitro using the USP Basket and Paddle Methods of USP Drug Release test of U.S. Pharmacopeia (2003) at 100 rpm in 900 ml aqueous buffer, is no greater than 30%. In other preferred embodiments, the difference, at any given time, between an amount of abusable drug released at one pH and an amount released at any other pH using the aforementioned methods is no greater than 50%, or no greater than 40%, or no greater than 35%, or no greater than 25%, or no greater than 20%, or no greater than 15%, or no greater than 10%, or no greater than 5%.
[00307] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said dosage forms of abusable drug providing in-vitro release rates by weight of between 0% to about 50% by weight of the abusable drug from the dosage form at one hour when measured by the USP Basket and Paddle Methods at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) at 37 °C. In other preferred embodiments, said release rate at one hour is between 0% to about 10% by weight, or 0% to about 20% by weight, or is between 0% to about 30% by weight, or 0% to about 40% by weight, or between 0% to about 60% by weight, or 0% to about 70% by weight, or 0% to about 80% by weight, or 0% to about 90% by weight, or 10% to about 50% by weight, or 10% to about 60% by weight, or 10% to about 70% by weight, or 10% to about 90% by weight, or 10% to about 100% by weight, or 30% to about 100% by weight, or 50% to about 100% by weight. [00308] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof and ADER, said dosage forms of abusable drug providing in- vitro release rates by weight of abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% to about 80% at 0.5 hours, and greater than about 40% at 1 hour. In other preferred embodiments, said release rate is between 0% to about 40% at 0.5 hours, and greater than about 60% at 1 hour; or between 0% to about 20% at 0.5 hours, and greater than about 40% at 1 hour; or between 0% to about 20% at 0.5 hours, and greater than about 20% at 1 hour; or between 0% to about 90% at 0.5 hours, and greater than about 60% at 1 hour; or between 0% to about 100% at 0.5 hours, and greater than about 60% at 1 hour; or between 0% to about 90% at 1 hour, and greater than about 40% at 2 hours; or between 0% to about 100% at 1 hour, and greater than about 60% at 2 hours; or between 0% to about 60% at 1 hour, and greater than about 40% at 2 hours; or between 0% to about 40% at 1 hour, and greater than about 30% at 2 hours; or between 0% to about 50% at 1 hour, and greater than about 40% at J 2 hours; or between 0% to about 30% at 1 hour, and greater than about 20% at 2 hours; or between 0% and about 50% at 1 hour, between about 0% and about 80% at 2 hours, between about 5% and about 100% at 4 hours and between about 10% and about 100% at 8 hours; or between 10% and about 60% at 1 hour, between about 15% and about 75% at 2 hours, between about 20% and about 95% at 4 hours and between about 30% and about 100% at 8 hours.
[00309] In some preferred embodiments, the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug . and (ii) ADER, such that the ratio of the mean Cmax of the abusable drug after single dose oral administration of the dosage form after tampering to the mean Cmax of abusable drug after single dose oral administration of an intact dosage form is not more than about 20: 1. In other embodiments of the invention, the mean Cmax ratio using the aforementioned test method is not more than about 15:1, or about 10:1, or about 7.5:1, or about 6:1, or about 5:1, or about 4:1, or about 3:1, or about 2:1, or about 1.5:1, or about 1.25:1.
[00310] In some preferred embodiments, the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean Cmax of the abusable drug after single dose oral administration of an immediate release reference product containing an equivalent amount of abusable drug to the mean Cmaχ of abusable drug after single dose oral administration of an intact dosage form of the invention is at least about 1.25:1. In other embodiments of the invention, the mean Cmax ratio using the aforementioned test method is at least about 1.5:1, or about 2:1, or about 3:1, or about 4:1, or about 5:1, or about 6:1, or about 10:1, or about 15:1 or about 20:1.
[00311] In some preferred embodiments, the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean AUC0-2 of the abusable drug after single dose oral administration of the dosage form after tampering to the mean AUC0-2 of abusable drug after single dose oral administration of an intact dosage form is not more than about 20:1. In other embodiments, the mean AUC ratio using the aforementioned test method is measured from time 0 to up to 1, 2.5, 3, 4, 5 or 6 hours post dose (i.e., AUCo-1, AUCo-2.5, AUC0-3, AUCo-4, AUC0-S and AUC0-6, respectively). In other embodiments of the invention, the mean AUCo-i, AUC0-2, AUCo-2.5, AUC0-3, AUC0-4, AUC0-5 and AUCo-6 ratios using the aforementioned test method are not more than about 15:1, or about 10:1, or about 7.5:1, or about 6:1, or about 5:1, or about 4:1, or about 3:1, or about 2:1 or about 1.5:1.
[00312] In some preferred embodiments, the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean AUCo-2 of the abusable drug after single dose oral administration of an immediate release reference product containing an equivalent amount of abusable drug to the mean AUC0-2 of abusable drug after single dose oral administration of an intact dosage form of the invention is at least about 1.25:1. In other embodiments, the mean AUC ratio using the aforementioned test method is measured from time 0 to up to 1, 2.5, 3, 4, 5 or 6 hours post dose (i.e., AUC0-I, AUCo-2.5, AUC0-3, AUC0-4, AUCo-5 and AUCo-6, respectively). In other embodiments of the invention, the mean AUC0-1, AUC0-2, AUCo-2.5, AUC0-3, AUC0-4, AUC0.5 and AUC0-6 ratios using the aforementioned test method are not more than about 15:1, or about 10:1, or about 7.5:1, or about 6:1, or about or about 5:1, or about 4:1, or about 3:1, or about 2:1 or about 1.5:1.
[00313] In some preferred embodiments, the oral dosage forni of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean Tmaχ of the abusable drug after single dose oral administration of the intact dosage form to the mean Tmax of abusable drug after single dose oral administration of an dosage form after tampering is not more than about 20:1. In other embodiments of the invention, the mean Tmaχ ratio using the aforementioned test method is not more than about 15:1, or not more than about 10:1, or not more than about 7.5:1, or not more than about 6:1, or not more than about 5:1, or not more than about 4:1, or not more than about 3:1, or not more than about 2:1, or not more than about 1.5:1, or not more than about 1.25:1.
[00314] In some preferred embodiments, the oral dosage form of the present invention is directed to an oral dosage form comprising: (i) an abusable drug and (ii) ADER, such that the ratio of the mean Tmax of the abusable drug after single dose oral administration of an immediate release reference product containing an equivalent amount of abusable drug to the mean Tmax of abusable drug after single dose oral administration of an intact dosage form of the invention is at least about 1.25:1. In other embodiments of the invention, the mean Tmax ratio using the aforementioned test method is at least about 1.5:1, or at least about 2: 1, or at least about 3:1, or at least about 4:1, or at least about 5:1, or at least about 6:1, or at least about 10:1, or at least about 15:1 or at least about 20:1.
[00315] In some preferred embodiments, the invention is directed to an oral dosage form comprising (i) an abusable drug and (ii) ADER, such that less than 70% of the abusable drug is released from the intact dosage form after 1 hour based on the in- vitro dissolution of the dosage form in 900 mL of 40% ethanol in water using the USP Basket and Paddle Methods at 50 rpm and 37°C. In other embodiments of the invention, the release rate of the abusable drug from the intact dosage form by the aforementioned USP basket method at 1 hours is 60% or less, 50% or less, 45% or less, 40% or less, 35% or less, 33% or less, 30% or less, 25% or less, 20% or less or 15% or less.
[00316] In certain preferred embodiments of the invention, the mean ratio of the amount of abusable drug released from the dosage form after mechanical tampering (e.g., after crushing with a single crush of a spatula or in the case of a capsule containing a solid, cutting into two pieces) to the amount of abusable drug released from the intact dosage form based on the dissolution at 0.5 hours of the dosage form in 900 mL of Simulated Gastric Fluid using the USP Basket and Paddle Methods at 50 rpm at 37 degrees °C is less than 20:1. In other embodiments of the invention, the mean ratio by the aforementioned USP basket method at 0.5 hours is 15:1 or less, 10:1 or less, 7.5:1 or less, 5:1 or less. 3:1 or less, 2:1 or less, 1.5:1 or less.
[00317] In certain preferred embodiments of the invention, the mean ratio of the amount of abusable drug released from the dosage form after mechanical tampering (e.g., after crushing with a single crush of a spatula or in the case of a capsule containing a solid, cutting into two pieces) to the amount of abusable drug released from the intact dosage form based on the dissolution at 1 hour of the dosage form in 900 mL of Simulated Gastric Fluid using the USP Basket and Paddle Methods at 50 rpm at 37 degrees 0C is less than 20:1. In other embodiments of the invention, the mean ratio by the aforementioned USP basket method at 1 hour is 15:1 or less, 10:1 or less, 7.5:1 or less, 5:1 or less. 3:1 or less, 2:1 or less, 1.5:1 or less.
[00318] In certain preferred embodiments of the invention, the mean ratio of the amount of abusable drug released from the dosage form after mechanical tampering (e.g., after crushing with a single crush of a spatula or in the case of a capsule containing a solid, cutting into two pieces) to the amount of abusable drug released from the intact dosage form based on the dissolution at 2 hours of the dosage form in 900 mL of Simulated Gastric Fluid using the USP Basket and Paddle Methods at 50 rpm at 37 degrees 0C is less than 20:1. In other embodiments of the invention, the mean ratio by the aforementioned USP basket method at 2 hours is 15:1 or less, 10:1 or less, 7.5:1 or less, 5:1 or less. 3:1 or less, 2:1 or less, 1.5:1 or less.
[00319] In some preferred embodiments, the present invention is directed to an oral dosage form comprising (i) an abusable drug and (ii) ADER, such that the ratio of the mean Cmax of the abusable drug after single dose oral administration of the dosage form after tampering to the mean Cmax of abusable drug after single dose oral administration of an intact dosage form is less than about 20:1. In other embodiments of the invention, said mean ratio using the aforementioned test method is less than about 15:1 or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 4:1, or less than about 3:1, or less than 2.5:1, or less than about 2:1, or less than about 1.75:1, or less than about 1.5:1, or less than about 1.25:1 or less than about 1.25:1
[00320] In some preferred embodiments, the present invention is directed to an oral dosage form comprising (i) an abusable drug and (ii) ADER, such that the ratio of the mean AUCo-2 of the abusable drug after single dose oral administration of an immediate release dosage form containing an equivalent amount of abusable drug to the mean AUCo-2 of abusable drug after single dose oral administration of an intact dosage form of the invention is at least 1.25:1. Li other embodiments of the invention, the mean AUC0-2 ratio using the aforementioned test method is at least about 1.5:1, or at least about 1.75:1, or at least about 2:1, or at least about 2.5:1, or at least about 3:1, or at least about 3.5:1, or at least about 4:1, or at least about 5:1, or at least about 6:1, or at least about 10:1 or at least about 15;1 or at least about 20:1.
[00321] The invention is also directed to methods of preventing abuse and misuse of an abusable drug utilizing the dosage forms disclosed herein. The method can comprise providing the abusable drug in an oral dosage form together with ADER, wherein the abusable drug is present in a form which is partially or substantially resistant to tampering (e.g., crushing, shear forces which break up the dosage form, solvent extraction, etc.). 100322] In certain preferred embodiments of the invention, the release for the abusable drug component of the formulation is expressed in terms of a ratio of the release achieved after tampering, relative to the amount released from the intact formulation. The ratio is therefore expressed as [Crushed]/[Whole], and it is desired that this ratio have a numerical range of not more than 20:1 (crushed release in 1 hour/intact release in 1 hour), based on in-vitro dissolution of the dosage form in 900 ml of Simulated Gastric Fluid using the USP Basket and Paddle Methods at 50 rpm and 37°C. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5 : 1 , or less than about 1.25 : 1.
[00323] In some embodiments, the abusable drug dosage form of the invention is bioequivalent when taken under fed and fasted conditions.
[00324] In some embodiments, the abusable drug dosage form of the invention upon administration provides a mean fed to fasted abusable drug AUCo-∞ difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%.
[00325] In some embodiments, the abusable drug dosage form of the invention upon administration provides a mean fed to fasted abusable drug Cm0x difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%. [00326] In some embodiments, the abusable drug dosage form of the invention is bioequivalent when taken under with and without 30 mL, 60 mL, 90 mL, 120 mL, 180 mL, 240 mL, 270 mL, 300 mL of a 40% ethanol solution.
[00327] In some embodiments, the abusable drug dosage form of the invention upon administration provides a mean alcohol to no alcohol state abusable drug AUCo-∞ difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%, said alcohol state induced by concurrent ingestion of the abusable drug with about 300 mL or about 270 mL, or about 240 mL, or about 210 mL, or about 180 mL or about 150 mL, or about 120 mL, or about 90 mL, or about 60 mL or about 30 mL of a 40% solution of ethanol.
[00328] In some embodiments, the abusable drug dosage form of the invention upon administration provides a mean alcohol to no alcohol state abusable drug Cmax difference of less than about 50%, or less than about 45%, or less than about 40%, or less than about 35%, or less than about 30%, or less than about 27%, or less than about 25%, or less than about 22%, or less than about 20%or less than about 18%, or less than about 15%, or less than about 12%, or less than about 10%, or less than about 8%, or less than about 6%, or less than about 5%, or less than about 3%, said alcohol state induced by concurrent ingestion of the abusable drug with about 300 mL or about 270 mL, or about 240 mL, or about 210 mL, or about 180 mL or about 150 mL, or about 120 mL, or about 90 mL, or about 60 mL or about 30 mL of a 40% solution of ethanol.
[00329] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof, ADER material, and a pharmacologic antagonist to the abusable drug. [00330] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof, ADER material, and a pharmacologic antagonist to a co-abused abusable drug, said co-abused abusable drug not part of the dosage form.
[00331] In some preferred embodiments, the dosage form provides an oral pharmaceutical composition comprising a therapeutically effective amount of abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof, ADER material, and a pharmacologic antagonist for both the abusable drug of the dosage form and a co-abused abusable drug, said co-abused abusable drug not part of the dosage form.
[00332] In some preferred embodiments, when the dosage form of the invention comprises a therapeutically effective amount of abusable drug or pharmaceutically acceptable salt thereof or mixture thereof, ADER material, and a pharmacologic antagonist to the abusable drug (and/or to a co-abused abusable drug, said co-abused abusable drug not part of the dosage form), some or all of the in vitro dissolution rates or in-vitro release rates (e.g., USP Basket and Paddle Methods) referred to in the embodiments and specifications herein and applicable to the abusable drug are also applicable to said antagonist. In the interest of brevity, such embodiments and specifications are not repeated here. In some preferred embodiments, the in vitro dissolution rates or in-vitro release rates for the abusable drug and the pharmacologic antagonists are the same. In other preferred embodiments, the in vitro dissolution rates or in-vitro release rates for the abusable drug and for the pharmacologic antagonists are from different.
[00333] In some preferred embodiments, when the dosage form of the invention comprises a therapeutically effective amount of abusable drug or pharmaceutically acceptable salt thereof or mixture thereof, ADER material, and an aversive agent, some or all of the in vitro dissolution rates or in-vitro release rates (e.g., USP Basket and Paddle Methods) referred to in the embodiments and specifications herein and applicable to the abusable drug are also applicable to said aversive agent. In the interest of brevity, such embodiments and specifications are not repeated here. In some preferred embodiments, the in vitro dissolution rates or in-vitro release rates for the abusable drug and the aversive agent are the same. In other preferred embodiments, the in vitro dissolution rates or in-vitro release rates for the abusable drug and for the aversive agent are different.
[00334] In some preferred embodiments, when the dosage form of the invention comprises a therapeutically effective amount of abusable drug or pharmaceutically acceptable salt thereof or mixture thereof, ADER material, and a pharmacologic antagonist to the abusable drug (and/or to a co-abused abusable drug, said co-abused abusable drug not part of the dosage form), some or all of the pharmacokinetic parameters (e.g., AUCo-t, AUCo--, AUCo- 8, AUC0-I2, AUC0-24, Cmax, C8, C12, C24, W or Tmax, Cmin, HVD, W50, steady state, percent fluctuation and AI) referred to in the embodiments and specifications herein and applicable to the abusable drug are also applicable to said antagonist. In the interest of brevity, such embodiments and specifications are not repeated here, hi some preferred embodiments, the pharmacokinetic parameters for the abusable drug and the pharmacologic antagonists are the same. In other preferred embodiments, the pharmacokinetic parameters for the abusable drug and for the pharmacologic antagonists are different.
[00335] In some preferred embodiments, the ADER material provides extended release of the abusable drug and the pharmacologic antagonist for the abusable drug, thereby significantly reducing the risk precipitating signs and symptoms of abusable drug withdrawal or an abstinence syndrome in abusable drug dependent and tolerant individuals, and diminution of the intended therapeutic response from the abusable drug. When such a dosage form is tampered with, the ADER material provides one line of defense and the antagonist provides yet another line of defense against drug abuse.
[00336] The amount of pharmacologic antagonist to the abusable drug in the oral dosage form will vary for a variety of reasons, including the choice of abusable drug, the potency of the abusable drug, the potency of the antagonist, the oral bioavailability of the abusable drug and the antagonist, the safety and tolerability of the antagonist, the degree of blockade sought to the effects of the abusable drug, the patients prior exposure to the abusable drug, the nature of the formulation (e.g., immediate release or extended release), the pharmacokinetics, pharmacodynamics and physicochemical characteristics of the abusable drug and the antagonist.
[00337] In some embodiments, pharmacologic antagonist to the abusable drug is about 0.00001 mg to 1000 mg, or about 0.00001 mg to about 700 mg, or about 0.001 mg to about 500 mg, or about 0.01 mg to about 400 mg, or about 0.1 mg to about 200 rag, or about 1 mg to about 100 mg, or about 0.00001 to about 500 mg, or about 0.001 to about 300 mg, or about 0.01 to about 200 mg.
[00338] In certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the time to confirmed perceptible pain relief after administration of the intact dosage form to the time to confirmed perceptible pain relief after administration of the tampered dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 15:1, .or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
[00339] In certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the time to meaningful pain relief after administration of the intact dosage form to the time to meaningful pain relief after administration of the tampered dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2: 1 , or less than about 1.5 : 1 , or less than about 1.25 : 1.
[00340] In certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the peak pain intensity difference score after administration of the tampered dosage form to the peak pain intensity difference score after administration of the intact dosage form is less than 10:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 8:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
[00341] hi certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the peak pain relief score after administration of the tampered dosage form to the peak pain relief score after administration of the intact dosage form is less than 10:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 8:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
[00342] In certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of change from baseline to two hours post-dose in pain intensity score after administration of the tampered dosage form to the change from baseline to two hours post-dose in pain intensity score after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
[00343] In certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the number of patients with pain who need to be treated to obtain > 50% pain relief in one patient (i.e., number needed to treat or NNT) after administration of the tampered dosage form to the NNT after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1. Preferably, the aforementioned at NNT is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post- dose. [00344] In certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the number needed to harm (referred to hereinafter as "NNH") due to moderate or severe nausea in healthy subjects (naive to said analgesic) after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post- dose.
[00345] ha certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the NNH due to moderate or severe sedation or drowsiness in healthy subjects (naϊve to said analgesic) after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is lessMhan about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00346] In certain preferred embodiments of the invention, wherein the abusable drug is an analgesic, the mean ratio of the NNH due to moderate or severe sedation or drowsiness in healthy subjects who are naϊve to said abusable drug and who are occasional or light consumers of alcohol after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1, said NNH measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 15:1, or less than about 10:1, or less than about 7:1, or less than about 5:1, or less than about 3:1, or less than about 2:1, or less than about 1.5:1, or less than about 1.25:1.
[00347] In certain preferred embodiments of the invention, wherein patients have the medical condition for which the abusable drug is medically used, the mean ratio of the number of patients who need to be treated (NNT) to obtain > 50% reduction in the cardinal sign or symptom of the medical condition (e.g., pain relief when the medical condition is pain, attention deficit hyperactivity disorder-rating scale [ADHD-RS-IV total scores] when the medical condition is ADHD, etc.) after administration of the tampered dosage form to the NNT after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNT is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00348] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to moderate or severe sedation or drowsiness in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00349] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to naive (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to moderate or severe sedation or drowsiness in healthy subjects who are naϊve to said abusable drug and who are occasional or light consumers of alcohol, after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1, said NNH measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1 , or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
[00350] In certain preferred embodiments of the invention, wherein the abusable drug is produces nausea on administration to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to moderate or severe nausea in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00351] In certain preferred embodiments of the invention, wherein the abusable drug produces CNS stimulation (e.g., the abusable drug is a stimulant, CNS-stimulant, psychostimulant, alkylxanthine or anorectic) upon administration to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of tachycardia (i.e., resting heart rate > 100 beats per minute) in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1 , or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00352] In certain preferred embodiments of the invention, wherein the abusable drug produces CNS stimulation (e.g., the abusable drug is a stimulant, CNS-stimulant, psychostimulant, alkylxanthine or anorectic) upon administration to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of palpitations in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16: 1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00353] In certain preferred embodiments of the invention, upon administration of abusable drug to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of headache in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1 , or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00354] In certain preferred embodiments of the invention, upon administration of abusable drug to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of dizziness in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15: 1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00355] In certain preferred embodiments of the invention, upon administration of abusable drug to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of lightheadedness in naive healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. hi other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1 :1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00356] In certain preferred embodiments of the invention, upon administration of abusable drug to naϊve {i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of dry mouth in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1 :1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00357] In certain preferred embodiments of the invention, upon administration of abusable drug to naϊve (i.e., abusable drug naive) healthy subjects, the mean ratio of the NNH due to the incidence of insomnia in naϊye healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
[00358] In certain preferred embodiments of the invention, upon administration of abusable drug to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of abdominal pain or abdominal discomfort in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1 , 1.5, 2, 3, 4, 5 or 6 hours post-dose
[00359] In certain preferred embodiments of the invention, upon administration of abusable drug to naive (i.e., abusable drug naϊve) healthy subjects, the mean ratio of the NNH due to the incidence of nervousness in naϊve healthy subjects after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 20:1. hi other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned at NNH is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00360] In certain preferred embodiments of the invention, upon administration of abusable drug to drug abusers and recreational drug users, the mean ratio of the drug liking score after administration of the tampered dosage form to the mean ratio of the drug liking score after administration of the intact dosage form is less than 20:1. hi other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose
[00361] In certain preferred embodiments of the invention, upon administration of abusable drug to drug abusers and recreational drug users, the mean ratio of the drug effect score after administration of the tampered dosage form to the mean ratio of the drug effect score after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1 , or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00362] In certain preferred embodiments of the invention, upon administration of abusable drug to drug abusers and recreational drug users, the mean ratio of the score on the "take again" questionnaire after administration of the tampered dosage form to the mean ratio of the score on the "take again" questionnaire after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00363] In certain preferred embodiments of the invention, upon administration of abusable drug to drug abusers and recreational drug users, the mean ratio of the score on the "coasting" questionnaire after administration of the tampered dosage form to the mean ratio of the score on the "coasting" questionnaire after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11 :1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00364] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to naive (i.e., abusable drug naive) healthy subjects, the mean ratio of impairment on the "critical tracking task" driving skills test in naϊve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00365] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to healthy subjects who are naive to said abusable drug and who are occasional' or light consumers of alcohol, the mean ratio of impairment on the "critical tracking task" driving skills test score in naϊve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1, said "critical tracking task" driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1 , or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
[00366] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of impairment on the "stop signal task" driving skills test in naϊve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1. In other -I l l-
embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00367] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to healthy subjects who are naϊve to said abusable drug and who are occasional or light consumers of alcohol, the mean ratio of impairment on the "stop signal task" driving skills test score in naϊve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1, said "stop signal task" driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%. m other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1 :1.
[00368] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to naϊve (i.e., abusable drug naϊve) healthy subjects, the mean ratio of impairment on the "Tower of London" driving skills test score in naϊve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1. Preferably, the aforementioned ratio is measured at 0.5, 1, 1.5, 2, 3, 4, 5 or 6 hours post-dose.
[00369] In certain preferred embodiments of the invention, wherein the abusable drug is produces sedation or drowsiness on administration to healthy subjects who are naϊve to said abusable drug and who are occasional or light consumers of alcohol, the mean ratio of impairment on the "Tower of London" driving skills test score in naϊve healthy subjects after administration of the tampered dosage form to the impairment after administration of the intact dosage form is less than 20:1 said "Tower of London" driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about L5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%. In other embodiments of the invention, the mean ratio using the aforementioned test method is less than about 18:1, or less than about 16:1, or less than about 15:1, or less than about 14:1, or less than about 12:1, or less than about 11:1, or less than about 10:1, or less than about 9:1, or less than about 8:1, or less than about 7:1, or less than about 6:1, or less than about 5:1, or less than about 4:1, less than about 3:1, or less than about 2.5:1, or less than about 2:1, less than about 1.75:1, or less than about 15:1, or less than about 1.25:1, or less than about 1.1:1.
[00370] The invention is also directed to a method of treating or preventing diseases and disorders amenable to treatment with abusable drugs. The method can comprise providing an oral dosage form containing an abusable drug and ADER, said dosage form an immediate release formulation, an extended release formulation or a formulation comprising both immediate release and extended release.
[00371] The oral dosage form containing an abusable drug in combination with
ADER includes, but is not limited to tablets or capsules. The dosage forms of the present invention may include any desired pharmaceutical excipients known to those skilled in the art. The oral dosage forms may further provide an immediate release of the abusable drug, hi certain preferred embodiments, the oral dosage forms of the present invention provide a sustained release of the abusable drug contained therein. Oral dosage forms providing sustained release of the abusable drug may be prepared in accordance with formulations/methods of manufacture known to those skilled in the art of pharmaceutical formulation.
[00372] The benefits of the abuse-resistant dosage form are especially great in connection with oral dosage forms of potent abusable drug, which can provide valuable therapeutic benefits but are prone to being abused. This is particularly true for sustained release abusable drug products which have a large dose of a desirable abusable drug intended to be released over a period of time in each dosage unit. Drug abusers take such sustained-release product and crush, grind, extract or otherwise damage the product so that the full contents of the dosage form become available for immediate absorption. Since such tampering of the dosage form of the invention results in the abusable drug also becoming available for absorption, the present invention provides a means for deterring such abuse. In addition, the present invention addresses the risk of overdose to non-abusing patients from "dumping" effect of the full dose of the abusable drug if the product is accidentally chewed or crushed, co-ingested with a significant amount of alcohol or taken interchangeably in fasted and fed states.
[00373] In certain preferred embodiments, a combination of two abusable drug is included in the formulation with the ADER. In further embodiments, one or more abusable drug and ADER are included and a further non-abusable drug is also included for the treatment of the same medical condition as the abusable drug or for the treatment of a different medical condition. [00374] Another embodiment of the invention is directed to a method of preventing or treating pain, addiction disorders, spasticity, musculoskeletal disorders, ADHD, insomnia, excessive sleepiness, daytime sleepiness, sleep disorders, anxiety disorders, panic attacks, agoraphobia, obsessive-compulsive disorders, psychiatric disorders, neurologic disorders, excess weight, obesity and other medical maladies responsive to treatment with the abusable drugs with the disclosed dosage forms. In certain preferred embodiments, the method of treating the aforementioned disorders in patients with a dosage form having less abuse potential comprises providing an oral dosage form containing an abusable drug and ADER; and orally administering the dosage form to provide a plasma level of abusable drug greater than the minimum therapeutic or minimum effective concentration of the abusable drug.
[00375] The invention is also directed to methods of preparing the dosage forms disclosed herein.
[00376] The benefits of the abuse-resistant dosage form are especially great in connection potent abusable drugs, which would provide valuable therapeutic benefits but would be prone to being abused. This is particularly true for oral dosage forms, including, in some preferred embodiments, sustained release dosage forms of abusable drugs which would have a large dose of a desirable drug intended to be released over a period of time in each dosage unit. Drug abusers may tamper the dosage form of the invention so that the full contents of the dosage form become available for immediate and maximal mood altering effects. The dosage form of the present invention would reduce the mood altering effects of the abusable drugs upon tampering and as such the invention provides pharmaceutical compositions, dosage forms and methods of deterring misuse, abuse, tampering and diversion of the dosage form.
[00377] In some embodiments of the invention, when the dosage form of the invention is tampered, the amount of abusable drug released in immediate release form is reduced, which in turn reduces the euphoric, pleasurable, reinforcing, rewarding, mood altering and toxic effects of the abusable drugs of the dosage form. [00378] When the dosage form of the present invention is orally administered as intended to humans, the abusable drugs is released into systemic circulation as intended and is therefore available for absorption into the body. However, if ' the dosage forms of the present invention is tampered (e.g., chemical, solvent, thermal or mechanical extraction, followed by administration into the body) the ADER of the invention would reduce the amount of abusable drugs available in immediate release form. Additionally, the dosage form of the invention substantially reduces the efficiency of drug aspiration into syringes, drug filtration after solvent extraction and drug extraction after attempts at chemical, mechanical or thermal extraction from both immediate and sustained release dosage form of the invention. These characteristic decrease the potential for abuse or diversion of the abusable drugs in the dosage form by blocking the mood altering, euphoric, pleasurable, reinforcing, rewarding or toxic effects of the abusable drug.
[00379] The term "tampering" or "tamper" means any manipulation by mechanical, thermal and/or chemical means which changes the physical or chemical properties of the dosage form, e.g., to liberate the abusable drugs for immediate release if it is in sustained release form, or to make the abusable drugs available for inappropriate use such as administration by an alternate route, e.g., parenterally. The tampering can be, e.g., by means of crushing, shearing, grinding, mechanical extraction, solvent extraction, solvent immersion, combustion, heating or any combination thereof.
[00380] The term "abuse", "drug abuse", "cannabinoid abuse", "stimulant abuse", "benzodiazepine abuse", or "sedative abuse", in the context of the present invention, when it refers to the effects of abusable drug in causing such, includes intermittent use, recreational use and chronic use of abusable drugs alone or in conjunction with other drugs means use: (i) in quantities or by methods and routes of administration that do not conform to standard medical practice; (ii) outside the scope of specific instructions for use provided by a qualified medical professional; (iii) outside the supervision of a qualified medical professional; (iv) outside the approved instructions on proper use provided by the drug's legal manufacturer; (v) which is not in specifically approved dosage forms for medical use as pharmaceutical agents; (vi) where there is an intense desire for and efforts to procure same; (vii) compulsive use; (viii) through acquisition by manipulation of the medical system, including falsification of medical history, symptom intensity, disease severity, patient identity, doctor shopping, prescription forgeries; (ix) where there is impaired control over use; (x) despite harm; (xi) by procurement from non-medical sources; (xii) by others through sale or diversion by the individual into the non-medical supply chain; (xiii) for medically unapproved or unintended mood altering purposes.
[00381] The term "mood altering" is defined for purposes of the present invention to mean that the "high", "liking", pleasurable, euphoric, alerting, calming, anxiolytic, auditory and visual perceptual alterations, relaxing, psychotomimetic, rewarding, reinforcing and toxic effects of the abusable drug.
[00382] The term "abuse resistant", "abuse deterrent", "tamper resistant",
"deter abuse" and "deter abuse" (as well of the words "resist" or "deter" when applied to abusable drugs of the invention) are used interchangeably in the context of the present invention and include pharmaceutical compositions and methods that resist, deter, discourage, diminish, delay and/or frustrate: (i) the intentional, unintentional or accidental physical or chemical manipulation or tampering of the dosage form (e.g., crushing, shearing, grinding, chewing, dissolving, melting, needle aspiration, inhalation, insufflation, extraction by mechanical, thermal and chemical means, and/or filtration); (ii) the intentional, unintentional or accidental use or misuse of the dosage form outside the scope of specific instructions for use provided by a qualified medical professional, outside the supervision of a qualified medical professional and outside the approved instructions on proper use provided by the drug's legal manufacturer (e.g., intravenous use, intranasal use, inhalational use and oral ingestion to provide high peak concentrations); (iii) the intentional, unintentional or accidental conversion of an extended release dosage form of the invention into a more immediate release form; (iv) the intentional and iatrogenic increase in physical and psychic effects sought by recreational drug users, addicts, and patients with pain who have an addiction disorder; (v) attempts at surreptitious administration of the dosage form to a third party (e.g., in a beverage); (vi) attempts to procure the dosage form by manipulation of the medical system and from non-medical sources; (vii) the sale or diversion of the dosage form into the non-medical supply chain and for medically unapproved or unintended mood altering purposes; (viii) the intentional, unintentional or accidental attempts at otherwise changing the physical, pharmaceutical, pharmacological and/or medical properties of the dosage form from what was intended by the manufacturer.
[00383] As used herein, the term "aversive agents", "aversion producing agents" and "aversive compounds" means to compounds contained within the dosage form that produce an aversive, undesirable, repugnant, distasteful, unpleasant, unacceptable physiologic or unacceptable psychic effects, or that pharmacologically block or reduce one or more of the following effects: mood alterations; euphoria, pleasure; a feeling of high; a feeling of drug liking; anxiolysis; mental stimulation; increased mental arousal; sedation; calmness; a state of relaxation; psychotomimesis; hallucinations; alterations in perception, cognition and mental focus; insomnia; hypersomnia; increased wakefulness or alertness; memory improvement; increased sexual gratification; increased sexual arousal; increased sexual desire and sexual anticipation; increased socialization; reduced social anxiety; psychologically reinforcement; and psychologically rewarding.
[00384] In some embodiments, the intention of the aversive agent is deter or further deter the misuse, abuse, tampering or diversion of the drug for use by addicts, drug abusers and recreational drug users. Preferably the aversive agent produces aversive effects only when the dosage form of the abusable drug of the invention is abused. - Preferably the aversive agent is contained within the dosage form at a dose or in a form that does not produce aversive effects when the dosage form of the abusable drug of the invention is taken as medically directed or in a manner that is consistent with the manufacturer's prescribing information, but which when abused or tampered with, produces an aversive effect. For example, in some embodiments, the dosage form of the abusable drug of the invention contains one or more aversive agents in a non- releasable form (i.e., sequestered) form, said aversive agent partially or substantially released upon tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like). For example, in some other embodiments, the dosage form of the abusable drug of the invention contains one or more aversive agents in releasable or partially releasable form, said dosage form not aversive when taken at medically approved doses or at doses consistent with the manufacturers prescribing information, said dosage form producing an aversive effect when taken in excess of medically approved doses or the manufacturers prescribing information. For example, in yet other embodiments, the dosage form of the abusable drug of the invention contains one or more aversive agents in a non-releasable form (i.e., sequestered) form, said aversive agent partially or substantially released upon tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like), and said aversive agent pharmacologically blocking the effects of the abusable drug and/or the effects of a co-abused drug, said co-abused drug not part of the dosage form of the invention.
[00385] As used herein, the term "ADER". "ADER material" and "ADER agent" refers to one or more compounds selected from the group consisting of: (a) hydrogenated Type I or Type II vegetable oils; (b) polyoxyethylene stearates and distearates; (c) glycerol monostearate; (d) poorly water soluble, high melting point (mp = 45 to 100° C) waxes, and mixtures thereof. In some preferred embodiments, ADER is a mixture of two or more compounds from the forgoing group [i.e., (a) to (d)]. In some preferred embodiments, to qualify as an "ADER" requires a mixture of two or more compounds from the form the foregoing group [i.e., (a) to (d)]. In some particularly preferred embodiments, to qualify as an "ADER" requires a mixture of two or more compounds selected from at least two categories[i.e., (a) to (d)].
[00386] In some preferred embodiments, references to the term "the invention",
"the present invention", "the pharmaceutical composition of the invention", "the dosage form of the invention", "the current invention" and embodiments of the invention in the embodiments, claims and specifications refer to pharmaceutical compositions, dosage forms, methods, processes and other innovations that comprise (i) one or more abusable drugs, in unsalified form or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof; (ii) one or more compounds selected from the group consisting of: (a) hydrogenated Type I or Type II vegetable oils; (b) polyoxyethylene stearates and distearates; (c) glycerol monostearate; (d) poorly water soluble, high melting point (mp = 45 to 100° C) waxes, and mixtures thereof, said compounds also referred to as ADER; and optionally [(Ui) and/or (iv)], (iii) other abusable or non-abusable drugs for the treatment of the same or a different medical condition; and/or (iv) pharmaceutical excipients, adjuvants and auxiliary agents including binders, disintegrants , fillers, diluents, anti- adherents or glidants, lubricants, stabilizers, wetting agents, pharmaceutically compatible carriers and dissolution rate modifiers, and channel and pore formers. ] hi some preferred embodiments, references to the term "the invention",
"the present invention", "the pharmaceutical composition of the invention", "the dosage form of the invention", "the current invention" and embodiments of the invention in the embodiments, claims and specifications refer to pharmaceutical compositions, dosage forms, methods, processes and other innovations that comprise (i) one or more abusable drugs, in unsalified form or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof; (ii) two or more compounds selected from the group consisting of: (a) hydrogenated Type I or Type II vegetable oils; (b) polyoxyethylene stearates and distearates; (c) glycerol monostearate; (d) poorly water soluble, high melting point (mp = 45 to 100° C) waxes, and mixtures thereof, said compounds also referred to as ADER; and optionally [(iii) and/or (iv)], (iii) other abusable or non-abusable drugs for the treatment of the same or a different medical condition; and/or (iv) pharmaceutical excipients, adjuvants and auxiliary agents including binders, disintegrants , fillers, diluents, anti- adherents or glidants, lubricants, stabilizers, wetting agents, pharmaceutically compatible carriers and dissolution rate modifiers, and channel and pore formers.
[00388] In some preferred embodiments, the dosage form may optionally also contain hydrophobic polymers, hydrophilic polymers, gums, protein derived materials, other waxes, shellac, other oils and mixtures thereof.
[00389] In some preferred embodiments, the invention is directed at an abusable drug dosage form, said dosage form having flotation capabilities to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage).
[00390] In some preferred embodiments, the invention is directed at an abusable drug dosage form, said dosage form having flotation capabilities to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage), said dosage form staying substantially afloat (i.e., floatable or buoyant) on a liquid surface for at least about 3 minutes. In other preferred embodiments, said dosage form stays substantially afloat for at least about 5 minutes, or at least about 7 minutes, or at least about 10 minutes, or at least about 15 minutes, or at least about 20 minutes, or at least about 25 minutes, or at least about 30 minutes, or at least about 40 minutes, or at least about 50 minutes, or at least about 60 minutes, or at least about 90 minutes, or at least about 120 minutes, or at least about 150 minutes, or at least about 180 minutes, or at least about 210 minutes, or at least about 240 minutes, or at least about 270 minutes, or at least about 300 minutes, or at least about 330 minutes, or at least about 360 minutes, or at least about 400 minutes.
[00391] In some preferred embodiments, the invention is directed at an abusable drug dosage form, said dosage form having a non-toxic dye to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage). [00392] In some preferred embodiments, the invention is directed at an abusable drug dosage form, said dosage form having a non-toxic bittering agent to deter surreptitious attempts at intoxication of another subject (e.g., in an alcoholic or non-alcoholic beverage).
[00393] In some preferred embodiments, the invention is directed at an abusable drug dosage form, said dosage form having a non-toxic bittering agent to deter oral or nasal ingestion of the dosage form.
[00394] In some preferred embodiments, the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined from first administration. In other preferred embodiments, the in vivo pharmacokinetic .parameters are derived or determined from steady state administration.
[00395] In some preferred embodiments, the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined under fed conditions. In other preferred embodiments, the in vivo pharmacokinetic parameters are derived or determined under fasted conditions.
[00396] In some preferred embodiments, the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined from an individual subject. In other preferred embodiments, the in vivo pharmacokinetic parameters are derived or determined from a population of subjects.
[00397] In some preferred embodiments, the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined in subjects having a Body Mass Index (BMI) between 18 and 26 kg/m2, inclusive (BMI = [weight in kg/ height in m2] x 10,000). In some preferred embodiments, the in vivo pharmacokinetic parameters of the specifications and claims are derived or determined in subjects having a Body Mass Index (BMI) > 38 kg/m2.
[00398] Also disclosed are methods for preventing and treating in pain, addiction disorders, spasticity, musculoskeletal disorders, ADHD, insomnia, excessive sleepiness, daytime sleepiness, sleep disorders, anxiety disorders, panic attacks, agoraphobia, obsessive-compulsive disorders, psychiatric disorders, neurologic disorders, excess weight, obesity and other medical maladies responsive to treatment with the abusable drugs in a human patient suffering from same, comprising a therapeutically effective amount of oral abusable drugs or pharmaceutically acceptable salts thereof or mixtures thereof.
[00399] All pain states are contemplated by this invention, regardless of etiology, mechanisms, duration, prior treatment response and anatomic location, including acute pain, inflammatory pain, chronic pain, cancer pain, visceral pain and neuropathic pain.
[00400] Also disclosed are methods of providing relief in a human patient suffering from neuropathic and chronic pain comprising a therapeutically effective amount of oral abusable drug which possesses analgesic properties or pharmaceutically acceptable salts thereof or mixtures thereof. In some preferred embodiments, the dosage form of the invention is intended for the treatment of neuropathic pain, peripheral neuropathic pain, central neuropathic pain, chronic pain, osteoarthritis, back pain, cancer pain, fibromyalgia, and chronic inflammatory pain.
[00401] Also disclosed are methods of providing relief in a human patient suffering from acute pain comprising a therapeutically effective amount of oral abusable drug which possesses analgesic properties or pharmaceutically acceptable salts thereof or mixtures thereof.
[00402] • All kinds of kits of the present invention are contemplated.
[00403] In some preferred embodiments, also provided are kits for use in treating or preventing the pain with the oral administration of an abusable drug or pharmaceutically acceptable salts thereof or mixtures thereof for a subject in need of such treatment, comprising: (i) a dosage form of the invention; (ii) a container for the dosage form; and optionally, any of (iii) to (vi): (iii) a container for individual units of the dosage form (e.g., individual tablets or capsules in blisters); (iv) educational instructions in any media about various medical conditions, including without limitation, pain, ADHD, sleep disorders, anxiety disorders, obesity, neurologic disorders and psychiatric disorders, their etiology, pathophysiology, consequences and treatment, including information on the potential for abuse and diversion and methods for prevention of same and information on the proper use and disposal of the medication; (v) containers or bags for the safe disposal of any used or remaining unused dosage form, preferably child proof and flushable; (vi) tamper evident and child proof packaging for the kit and its contents.
[00404] The amount of abusable drug in the oral dosage form will vary depending on variety of physiologic, pharmacologic, pharmacokinetic, pharmaceutical and physicochemical factors, including: (i) the choice of abusable drug as the unsalified form, pharmaceutically acceptable salt or mixtures therof; (ii) the nature of the oral dosage form (e.g, immediate release or extended release); (iii) the intensity and intractability of the medical condition; (iv) the absorption, metabolism, distribution and excretion of orally administered abusable drug in healthy subjects and in patients with various diseases and disorders, including renal and hepatic impairment; (v) the presence of comorbid pathology; (vi) the patient's risk of iatrogenic side effects; (vii) the tolerability of the dose, including the patient's propensity for abusable drug associated side effects; (viii) use of other drugs to treat the same medical condition; (ix) the efficiency of the dosage form; (x) the physicochemical properties of the abusable drug, including its solubility and hydrophilicity.
[00405] The invention is also directed to methods of preparing the dosage forms disclosed herein.
[00406] In certain preferred embodiments, the abusable drug in the dosage form is combined with one or more other drugs for the treatment of the same medical condition as the abusable drug or for the treatment of a different medical condition. All modes of co-administration are contemplated, including via an oral, subcutaneous, direct intravenous, slow intravenous infusion, continuous intravenous infusion, intravenous or epidural patient controlled analgesia (PCA and PCEA), intramuscular, intrathecal, epidural, intracisternal, intramuscular, intraperitoneal, transdermal, topical, transmucosal, buccal, sublingual, transmucosal, inhalation, intranasal, epidural, intra-atricular, intranasal, rectal or ocular routes. [00407] The term "first administration" means administration of a dose of the present invention at the initiation of therapy to an individual patient or a patient population.
[00408] The term "steady state" means that the amount of the drug reaching the system is approximately the same as the amount of the drug leaving the system. Thus, at "steady-state", the patient's body eliminates the drug at approximately the same rate that the drug becomes available to the patient's system through absorption into the blood stream.
[00409] As used herein the terms: (i) "AUCo-t" means area under the plasma drug concentration-time curve from time zero to the "t", where t is the time point of the maximum intended dosing frequency of the dosage form (e.g., 4 hours, 6 hours, 8 hours, 12 hours or 24 hours for dosage forms intended to be administered every 4 hours, every 6 hours, every 8 hours, every 12 hours and every 24 hours, respectively, thereby providing an AUCo-t time interval of 0 to 4 hours, 0 to 6 hours, 0 to 8 hours, 0 to 12 hours and 0 to 24 hours, respectively); (ii) "AUCo-~" means area under the plasma drug concentration- time curve from time zero to infinity; (iii) "AUCo-s" means area under the plasma drug concentration-time curve from time zero to 8 hours after dosing; (iv) "AUCo-i 2" means area under the plasma drug concentration-time curve from time zero to 12 hours after dosing; (v) "AUCo-24" means area under the plasma drug concentration-time curve from time zero to 24 hours after dosing; (vi) "Cmax" means the maximum observed plasma drug concentration; (vii) "C8" means the plasma drug concentration at 8 hours after dosing; (viii) "C n" means the plasma drug concentration at 12 hours after dosing; (ix) "C24" means the plasma drug concentration at 24 hours after dosing; (x) "tmax" or "Tmax" means the time of the observed maximum drug concentration (also known as the time at which Cm3x occurs); (xi) "Cmin" means the minimum observed drug concentration following the maximum plasma concentration or the concentration at the end of the intended dosing interval; (xii) "time at which Cmin occurs" means the time at when the minimum observed drug concentration occurs; (xiϋ) "half value duration" or "HVD" means the duration over the dosing interval during which plasma concentration of drug are greater than or equal to one-half of Cmax, obtained by calculating the time interval beginning when the interpolated concentration first equals or exceeds one-half of Cmax and ending at the first time point for which the interpolated concentration falls below one-half of Cmax; (xiv) "Ws0" means the duration of the dosing interval over which the plasma concentrations are equal to or greater than 50% of the peak concentration; (xv) "steady state" is a state of equilibrium wherein the amount of the drug reaching the system is approximately the same as the amount of the drug leaving the system or put another way, the patient's body eliminates the drug at approximately the same rate that the drug becomes available to the patient's system through absorption into the blood stream, said "time to steady state" measured by calculating the Cmin after each sequential dosing of drug administered at the intended dosing frequency until two consecutive Cmin's are not statistically different at a 10% significance level (p = 0.10); (xvi) "percent fluctuation" means the variation in plasma concentrations of the drug computed as: (a) (Cmaχ-Cmjn)/Cmjn x 100 (for an individual patient) and (mean Cmax-mean Cmjn)/mean Cmin x 100 (for a population); or (b) (Cmax-Cmin)/Cav x 100 (for an individual patient) and (mean Cmax-niean Cmin)/mean Cav x 100 (for a population); (xvii) "accumulation index" or "AI" means the ratio of the plasma concentration of the drug at the end of the intended dosing interval (i.e., 8 hours for a Q8H dosage form, 12 hours for a Ql 2H dosage form, and 24 hours for a Q24H dosage form) after administration, determined at steady-state (Cssmin) to the plasma concentration of the drug at the end of the intended dosing interval determined at first administration (i.e., after the first dose).
[00410] Pharmacokinetic parameters of the invention are be computed from first administration and steady state pharmacokinetic studies conducted in an individual subject or in a population of subjects in the fasted or fed states. The AI and percent of steady state computations requires both single dose (i.e., first administration) and steady state pharmacokinetic assessment.
[00411] In certain preferred embodiments of the present invention, an effective amount of abusable drug in immediate release form is included in the controlled release unit dose abusable drug formulation to be administered. The immediate release form of the abusable drug is preferably included in an amount which is effective to shorten the time to Cmax or increase the magnitude of the Cmax of the abusable drug in the blood (e.g., plasma). In such embodiments, an effective amount of the abusable drug in immediate release form may be coated onto the substrates of the present invention. For example, where the extended release abusable drug from the formulation is due to a controlled release coating, the immediate release layer would be overcoated on top of the controlled release coating. On the other hand, the immediate release layer maybe coated onto the surface of substrates wherein the abusable drug is incorporated in a controlled release matrix. Where a plurality of the sustained release substrates comprising an effective unit dose of the abusable drug are incorporated into a hard gelatin capsule, the immediate release portion of the abusable drug dose may be incorporated into the gelatin capsule via inclusion of the sufficient amount of immediate release abusable drug as a powder or granulate within the capsule. Alternatively, the gelatin capsule itself may be coated with an immediate release layer of the abusable drug. In some other embodiments, the immediate release abusable drug is in liquid form, for example as a capsule within a capsule or as a liquid in contact with an extended release dosage form within a capsule. One skilled in the art would recognize still other alternative manners of incorporating the immediate release abusable drug into the unit dose. Such alternatives are deemed to be encompassed by the appended claims. By including such an effective amount of immediate release abusable drug in the unit dose, they may experience of relatively higher levels of symptom relief or faster symptom relief.
[00412] In certain preferred embodiments, any one or all of the above in- vivo parameters are achieved after a first administration (often referred to as "single dose administration") of the dosage form to a human patient or a population of human patients.
[00413] In certain alternative embodiments, any one or all of the above in-vivo parameters are achieved after steady state administration of the dosage form to a human patient or a population of human patients. [00414] Perceptible Pain Relief, Confirmed Perceptible Pain Relief and
Meaningful Pain Relief are assessed and defined as follows: At the time of dosing with the study medication, a trained member of study staff starts two stopwatches for each patient. The patient is instructed to stop the first stopwatch at the time of perceptible pain relief and the second stopwatch at the time when they first experience meaningful pain relief. The usual definitions of the perceptible and meaningful pain relief are as follows: Perceptible Pain Relief is when the patient begins to feel any pain relieving effect from the drug. The patient is typically instructed as follows: "I would like you to stop the first stopwatch when you first feel any pain relief whatsoever. This does not mean you feel completely better, although you might, but when you first feel any difference in the pain that you have had". Meaningful Pain Relief is when the patient feels their pain relief is meaningful to them. The patient is typically instructed as follows: "I would like you to stop the second stopwatch when you have meaningful pain relief. That is, when the relief from the pain is meaningful to you". Confirmed Perceptible Pain Relief is Perceptible Pain Relief in those patients who go on to also have Meaningful Pain Relief.
[00415] As used herein, "NNT" or "the number needed to treat" is the number of patients who need to be treated in order for one patient to obtain > 50% reduction in signs or symptoms (e.g., > 50% reduction in pain intensity or ADHD-RS-IV score).
[00416] The "NNH" or "number needed to harm" is a measure that indicates how many patients would require a specific treatment to cause harm in one patient. As used herein, the "NNH or "number needed to harm" is a measure that indicates: (i) how many abusable drug naive healthy subjects would require treatment to cause moderate or severe sedation (or drowsiness) in one subject, where moderate to severe sedation or drowsiness is defined as a VAS score of > 50 mm on a 100 mm scale bounded on the left by "no sedation or drowsiness" and on the right by "extreme sedation or drowsiness"; (ii) how many abusable drug naive healthy subjects would require treatment to cause moderate or severe nausea in one subject, where moderate to severe nausea is defined as a VAS score of > 50 mm on a 100 mm scale bounded on the left by "no nausea" and on the right by "extreme nausea"; (iii) how many abusable drug naϊve healthy subjects would require treatment to cause tachycardia in one patient, where tachycardia is defined as a heart rate at rest which is > 100 beats per minute; (iv) how many abusable drug naϊve healthy subjects would require treatment to cause palpitations in one subject, where palpitation is defined as a sensation of rapid or irregular heartbeats; (v) how many abusable drug naϊve healthy subjects would require treatment to cause headache in one subject, where headache is defined as pain in the head; (vi) how many abusable drug naϊve healthy subjects would require treatment to cause dizziness in one subject, where dizziness is defined as unsteadiness, imbalance, lightheadedness, spinning sensation or sensation that one is falling; (vii) how many abusable drug naϊve healthy subjects would require treatment to cause a sensation of dry mouth in one subject, where dry mouth is defined as abnormal dryness of the mouth associated with decreased secretion of saliva; (viii) how many abusable drug naϊve healthy subjects would require treatment to cause insomnia in one subject, where insomnia is defined as difficulty sleeping (e.g., inability to fall asleep or remain asleep); (ix) how many abusable drug naϊve healthy subjects would require treatment to cause abdominal pain or discomfort in one subject, where abdominal pain or discomfort is defined as pain in the stomach region or belly; and/or (x) how many abusable drug naϊve healthy subjects would require treatment to cause nervousness in one subject, where nervousness is defined as nervousness, jitteriness or an unpleasant, emotional state of high energy. ] The "drug effects" questionnaire assesses the extent to which subjects currently felt a drug effect, on a scale of 1 to 5 (1 = "I feel no effect from it at all"; 2 = "I think I feel a mild effect, but I'm not sure"; 3 = "I feel an effect, but it is not real strong"; 4 = "I feel a strong effect"; 5 = "I feel a very strong effect"). This questionnaire can be used to examine the overall drug effects of abusable drugs given intact and upon tampering, preferably in drug abusers and recreational drug users without the medical condition for which the drug is effective. [00418] The "drug liking" questionnaire assesses the extent to which subjects currently like the effects of the drug on a 100-mm VAS, bounded on the left by "0 = dislike a lot", bounded on the right by "100 = like a lot". This questionnaire can be used to examine the overall drug liking of abusable drugs given intact and upon tampering, preferably in drug abusers and recreational drug users without the medical condition for which the drug is effective.
[00419] The "take again" questionnaire assesses whether subjects would take the abusable drug again if given the opportunity. The patient is asked "If given an opportunity, would you take this drug again? (circle one: YES or NO). This questionnaire can be used to examine the overall desirability of the drug experience with the abusable drugs taken intact and taken after . tampering, preferably in drug abusers and recreational drug users without the medical condition for which the drug is effective.
[00420] On the "coasting" questionnaire the patient is asked to put a mark on a horizontal line that best describes their response to the question: "Do you feel like you are coasting or spaced out? The horizontal line is a visual analog scale (VAS) bounded on the left by "not at all" and on the right by "extremely". This questionnaire can be used to examine the degree to which subjects feel like they are coasting or spaced out with the abusable drugs taken intact and taken after tampering, preferably in drug abusers and recreational drug users without the medical condition for which the drug is effective.
[00421] Three performance tasks may be employed for measuring skills related to driving.
[00422] The "critical tracking task" measures the patient's ability to control a displayed error signal in a first-order compensatory tracking task. The error is displayed as a horizontal deviation of a cursor from the midpoint on a horizontal, linear scale. Compensatory joystick movements correct the error by returning the cursor to the midpoint. The frequency at which the patient loses the control is the critical frequency. The critical tracking task measures the psychomotor control during a closed loop operation. It is a laboratory analog to on-the-road tracking performance. [00423] The "stop signal task" measures motor impulsivity, which is defined as the inability to inhibit an activated or pre-cued response leading to errors of commission. The task requires patients to make quick key responses to visual go signals, i.e. the letters ABCD presented one at a time in the middle of the screen, and to inhibit any response when a visual stop signal, i.e. "*" in one of the four corners of the screen, is presented at predefined delays. The main dependent variable is the stop reaction time on stop signal trials that represents the estimated mean time required to inhibit a response.
[00424] The Tower of London (TOL) is a decision-making task that measures executive function and planning. The task consists of computer generated images of begin- and end-arrangements of three colored balls on three sticks. The subject's task is to determine as quickly as possible, whether the end- arrangement can be accomplished by "moving" the balls in two to five steps from the beginning arrangement by pushing the corresponding number coded button. The total number of correct decisions is the main performance measure.
[00425] For the purposes of in vivo testing, unless specified otherwise, pain intensity is measured on a VAS or categorical scale. On the categorical scale, the patient is asked "My pain at this time is: None = 0, Mild = 1, Moderate = 2, Severe = 3. On the VAS, the patient is asked "My pain at this time is" (with VAS anchors: "No Pain" and "Extreme Pain").
[00426] For the purposes of in vivo testing, unless specified otherwise, pain relief is measured on a categorical scale. The patient is asked "My relief from starting pain is: None = 0, A little = 1, Some = 2, A lot = 3, Complete = 4.
[00427] In certain preferred embodiments, the amount of abusable drug in the dosage form is about 0.01 μg to 1500 mg. In other more preferred embodiments, the amount of abusable drug in the dosage form is about 0.1 μg to 1000 mg or about 0.1 μg to 1500 mg. In most preferred embodiments, the amount of abusable drug in the dosage form is about 0.01 μg to 750 mg, or about 0.01 μg to about 500 mg, or about 0.01 μg to about 250 mg, or about 0.1 μg to about 500 mg, or 0.1 μg to about 250, or about 0.1 μg to about 250 mg, or about 1 μg to about 1500 mg, or 1 μg to about 1000 mg, or about 1 μg to about 100 mg, or about 5 μg to about 1500 mg, or about 5 μg to about 1000 mg, or about 5 μg to about 500 mg, or about 10 μg to about 1000 mg, or about 10 μg to about 500 mg, or about 100 μg to about 1000 mg, or about 1 mg to about 1500 mg, or about 1 mg to about 1000 mg, or about 1 mg μg to about 800 mg, or about 1 mg to about 500 mg.
[00428] In certain embodiments, the amount of ADER in the claimed composition may be about 1 mg to 1500 mg. In a preferred embodiment, the amount of ADER in the claimed composition may be about 10 mg to 800 mg. In a most preferred embodiment, the amount of ADER in the claimed composition may be about 50 mg to 600 mg.
[00429] In certain preferred embodiments of the present invention, the ratio of the abusable drug and the ADER is about 1:10,000 to about 10,000:1 by weight, preferably about 1:1000 to about 1000:1 by weight, more preferably 1 :250 to 250:1.
[00430] The term "USP Basket and Paddle Methods" is the Basket and Paddle
Method described, e.g., in specified in the United States Pharmacopeia, USP- 28 NF-23 (2005), published by the United States Pharmacopeial Convention, Inc, and herein incorporated by reference.
[00431] The term "pH-dependent" for purposes of the present invention is defined as having characteristics (e.g., dissolution) which vary according to environmental pH.
[00432] The term "pH-independent" for purposes of the present invention is defined as having characteristics (e.g., dissolution) which are substantially unaffected by pH.
[00433] The term "bioavailability" is defined for purposes of the present invention as the rate and extent to which the drug (e.g., the abusable drug) is absorbed from the unit dosage forms.
[00434] As used herein with respect to the abusable drug dosage forms of the invention, the term "oral", "oral dosage form", "oral pharmaceutical dosage form", "oral administration", and "oral route", refer to any method of administration involving contact with the mouth and oral mucosa, including the ingestion of intact drugs (e.g., capsules, tablets, liquids swallowed whole), lingual, sublingual administration, buccal administration and transmucosal administration. Particularly preferred embodiments involve oral ingestion of intact drugs (e.g., capsules, tablets, liquids swallowed whole).
[00435] AU oral pharmaceutical dosage forms of the invention are contemplated, including oral suspensions, tablets, capsules, lozenges, effervescent tablets, effervescent powders, powders, solutions, powders for reconstitution, transmucosal films, buccal products, oral mucoretentive products, oral gastroretentive tablets and capsules, orally disintegrating tablets, fast dissolving tablets, fast dispersing tablets, fast disintegrating dosage forms, administered as immediate release, delayed release, modified release, enteric coated, sustained release, controlled release, pulsatile release and extended release dosage form.
[00436] As used herein, "controlled release" is interchangeable with "extended release", "sustained release", "modified release", "delayed release" and the like. Such products provide a longer duration of action than conventional immediate release formulations of the same drugs and are usually administered every 8, 12 or 24 hours.
[00437] Controlled release dosage forms of the present invention release abusable drug from the oral dosage form at slower rate than immediate release formulations. In some preferred embodiments, controlled release dosage forms release abusable drug at such a rate that blood (e.g., plasma) concentrations (levels) or therapeutic effects are maintained within the therapeutic range (above the minimum effective therapeutic concentration) but below toxic levels for intended duration (e.g., over a period of 1 to 24 hours, preferably over a period of time indicative of Q4, Q6, QS, Q12 or Q24H administration). Notwithstanding the foregoing, in some preferred embodiments, the controlled release formulations of the present invention provide therapeutic effects for a duration that is longer or substantially longer than the duration of meaningful or detectable plasma concentrations of abusable drug. Controlled release dosage forms may be administered around the clock on a scheduled or time contingent basis, or on an as needed or PRN basis, e.g., Q3 PRN, Q4 PRN, Q6 PRN, Q8 PRN, Q12 PRN or Q24H PRN administration.
[00438] The term "immediate release abusable drug" for purposes of the present invention is abusable drug for oral administration in a dosage form which formulated to release the active drug from the dosage form immediately (i.e., without an attempt to delay or prolong the release of the active drug from the dosage form as is the case for extended release dosage forms). In the absence of a commercially available oral immediate release abusable drug product, an available parenteral formulation of abusable drug or a salt thereof may be used orally or a solution of abusable drug or a salt thereof may be prepared for the purpose of in vivo testing requiring immediate release abusable drug.
[00439] For purposes of the invention, the controlled release formulations disclosed herein and the immediate release control formulations are dose proportional. In such formulations, the pharmacokinetic parameters (e.g., AUC and Cmax) increase linearly from one dosage strength to another. Therefore the pharmacokinetic parameters of a particular dose can be inferred from the parameters of a different dose of the same formulation.
[00440] The phrase "cardinal sign or symptom" and "cardinal sign or symptom of said medical condition" when referring to the use of the abusable drug of the present invention means the major sign or symptom of the medical condition for which the abusable drug is approved or commonly used, said sign or symptom commonly used as the primary endpoint in clinical trials published in peer-review journals and for approval of the drug by the U.S. FDA and other major drug regulatory authorities (e.g., the EMEA).
[00441] The term "agonist" means a ligand that binds to a receptor and alters the receptor state resulting in a biological response. Conventional agonists increase receptor activity, whereas inverse agonists reduce it (See Neubig et al, IUPHAR Committee on Receptor Nomenclature and Classification, Pharmacol Rev, 2003; Howlett et al., MoI Pharmacol, 1988). [00442] The term "opioid agonist" means a molecule that causes a specific physiologic, pathophysiologic or pharmacologic effect after binding to an opioid receptor.
[00443] An "antagonist" is a drug or ligand that reduces the action of another drug or ligand, generally an agonist. Many antagonists act at the same receptor macromolecule as the agonist. (See Neubig et al, IUPHAR Committee on Receptor Nomenclature and Classification, Pharmacol Rev, 2003; Howlett et al., MoI Pharmacol, 1988).
[00444] The term "receptor" means a molecule within a cell, on a cell surface, on a membrane, in tissue, in fluid or otherwise found in humans that serve as a recognition or binding site to cause specific physiologic, pathophysiologic or pharmacologic effects. The term "receptor" also means a cellular macromolecule, or an assembly of macromolecules, that is concerned directly and specifically in chemical signaling between and within cells. Combination of a hormone, neurotransmitter, drug, ligand, or intracellular messenger with its receptor(s) initiates a change in cell function (Neubig et al, IUPHAR Committee on Receptor Nomenclature and Classification, Pharmacol Rev, 2003).
[00445] The term "abuse", "drug abuse", "cannabinoid abuse",
"benzodiazepine abuse", "barbiturate abuse", "methylphenidate abuse", "stimulant abuse" or "psychostimulant abuse" in the context of the present invention includes intermittent use, recreational use and chronic use of abusable drugs alone or in conjunction with other drugs: (i) in quantities or by methods and routes of administration that do not conform to standard medical practice; (ii) outside the scope of specific instructions for use provided by a qualified medical professional; (iii) outside the supervision of a qualified medical professional; (iv) outside the approved instructions on proper use provided by the drug's legal manufacturer; (v) which is not in specifically approved dosage forms for medical use as pharmaceutical agents; (vi) where there is an intense desire for and efforts to procure same; (vii) with evidence of compulsive use; (viii) through acquisition by manipulation of the medical system, including falsification of medical history, symptom intensity, disease severity, patient identity, doctor shopping, prescription forgeries; (ix) where there is impaired control over use; (x) despite harm; (xi) by procurement from non-medical sources; (xii) by others through sale or diversion by the individual into the non-medical supply chain; (xiii) for medically unapproved or unintended mood altering purposes.
[00446] As used herein, "abusable drugs", "abusable drug", "abusable pharmaceuticals", "abusable substances" and "abusable dosage forms" are: (i) substances which have medical applications for the prevention or treatment of diseases and disorders (i.e., "therapeutically active agents"); and (ii) substances which have the potential for being abused by drug abusers, recreational drug users and individuals with an addiction disorder for intermittent use, recreational use or chronic use, wherein the abusable drug is being abused for one or more of the following effects: mood alterations; euphoria, pleasure; a feeling of high; a feeling of drug liking; anxiolysis; mental stimulation; increased mental arousal; sedation; calmness; a state of relaxation; psychotomimesis; hallucinations; alterations in perception, cognition and mental focus; insomnia; hypersomnia; increased wakefulness or alertness; memory improvement; increased sexual gratification; increased sexual arousal; increased sexual desire and sexual anticipation; increased socialization; reduced social anxiety; psychologically reinforcement; and psychologically rewarding. It reasons that various abusable drugs of the invention will have different abusable characteristics being sought after by the drug addict or recreational drug user. Abusable drugs of the invention specifically exclude all opioid agonists.
(00447] Representative example of abusable drugs are, without limitation amphetamine and amphetamine like CNS-stimulants, amphetamine analogs, CNS-stimulants, nicotine, methylphenidate, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, non-benzodiazepine hypnotics and CNS depressants, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, hypnotics and psychoactive agents, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and prodrugs, analogs or derivative thereof, but specifically excluding all opioid agonists.
[00448] In some embodiments, as used herein, the phrase "abusable drugs",
"abusable drug", "abusable pharmaceuticals", "abusable substances" and "abusable dosage forms" refer only to methylphenidate, or only to amphetamine analogs, or only to amphetamines and amphetamine analogs, or only to modafinil, or only to CNS or psychostimulants, or only to cannabinoid agonists, or only to nicotine, or only to alkylxanthines, or only to anorectics, or only to barbiturates, or only to benzodiazepine agonists, or only to non- benzodiazepine hypnotics and CNS depressants, or only to anxiolytic agents, or only to sedatives, or only to hypnotics, or only to controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended), or only to drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and their respective pharmaceutically acceptable salts, prodrugs or analogs in racemic or enantiomeric form.
[00449] In other embodiments, as used herein, the phrase "abusable drugs",
"abusable drug", "abusable pharmaceuticals", "abusable substances" and "abusable dosage forms" refer to one or more compounds selected from the group comprising methylphenidate, amphetamine analogs, amphetamines, modafinil, CNS or psychostimulants, cannabinoid agonists, nicotine, alkylxanthines, anorectics, barbiturates, benzodiazepine agonists, non- benzodiazepine hypnotics, CNS depressants, anxiolytics, sedatives, hypnotics, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended), and drugs listed under the United States Psychotropic Substances Act of 1978, as amended, and their respective pharmaceutically acceptable salts, prodrugs or analogs in racemic or enantiomeric form.
[00450] In some preferred embodiments, abusable drugs of the invention may be in unsalified form as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof. In other preferred embodiment, abusable drugs of the invention may be pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymotphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof. In particularly preferred embodiment, abusable drugs of the invention may be in unsalified form, pharmaceutically acceptable salts, prodrugs, esters or with a covalently bound pharmaceutically active moiety or mixtures thereof, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof.
[00451] A number of assays are available to determine whether a drug is a benzodiazepine agonist or benzodiazepine antagonist, using in vivo and in vitro bioassay systems.
[00452] Benzodiazepine agonist are known or readily determined by individuals who practice the art. All benzodiazepine agonists are contemplated by the invention, including benzodiazepine BZl (omega 1) receptor agonists. Preferably, the benzodiazepine agonist useful for the present invention may be selected from the group consisting of alprazolam, bromazepam, brotizolam, camazepam, chlordiazepoxide, cinolazepam, clobazam, clonazepam, clorazepate, desalkylflurazepam, diazepam, estazolam, flunitrazepam, flurazepam, halazepam, ketazolam, loprazolam, lorazepam, lormetazepam, medazepam, metaclazepam, midazolam, nitrazepam, nordazepam, oxazepam, phenazepam, pinazepam, prazepam, quazepam, temazepam, tetrazepam, triazolam, zaleplone, Zolpidem and zopiclone.
[00453] The term "benzodiazepine agonist" means a substance that binds to one or more benzodiazepine receptors or recognition sites, their subtypes and splice variants to exert an agonist or partial agonist effect.
[00454] For the purposes of the present invention, the term "benzodiazepine receptor" or "benzodiazepine recognition site" includes one or more benzodiazepine receptors or recognition sites, the benzodiazepine BZl (omega 1) receptor, the gamma-aminobutyric acid (GABA)-benzodiazepine receptor complex, the gamma-aminobutyric acid type A (GABAA) receptor and their respective subtypes and splice variants.
[00455] Non-benzodiazepine hypnotics and CNS depressants are known or readily determined by individuals who practice the art. Non-benzodiazepine hypnotics and CNS depressants include sodium oxybate, diphenhydramine, chlorpheniramine, trazadone, amitriptyline, cyclobenzaprine, methocarbamol, carisoprodol, ramelteon, and drugs from the barbiturate, antihistamine, antidepressant and melatonin receptor agonist class.
[00456] Cannabinoid agonists are known or readily determined by individuals who practice the art. Preferably, the cannabinoid agonist useful for the present invention may be selected from the group consisting of inhibitors of cannabinoid agonist metabolism (e.g., without limitation, URB602, an inhibitor of monoacylglycerol lipase which catalyzes 2-arachidonoylglycerol hydrolysis) THC, nabilone, dronabinol, cannabidiol, 9-THC propyl analog, cannabidiol, cannabidiol propyl analog, carmabinol, cannabichromene, cannabichromene propyl analog, cannabigerol, cannabinoid terpenoids, cannabinoid flavonoids, endocannabinoids, anandamide, (R)- methanandamide,and 2-arachidonoylglycerol, THC-like ABC tricyclic cannabinoid analogues, exemplified by HU210 and desacetyllevonantradol; synthetic AC bicyclic and ACD tricyclic cannabinoid analogues, exemplified by CP55940, and CP55244 and aminoalkylindole compounds, exemplified by WIN55212-2, and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof. (Little et al., Pharmacol. Biochem. Behav, 1989;Howlett et al., Neuropharmacology, 1990;Johnson et al, In: Cannabinoids as Therapeutic Agents (Mechoulam, R., ed.), CRC Press, 1986; Howlett et al., MoI Pharmacol, 1988; D'Ambra et al., J Med Chem, 1992; Pacheco et al., J Pharmacol Exp Ther, 1991; Compton et al, J Pharmacol Exp Ther, 1992; Howlett et al, Pharmacol Rev, 2002; Fowler. Fundam Clin Pharmacol. 2006;20:549-62; Karanian and Bahr, Curr MoI Med 2006;6:677- 84; Singh and Budhiraja, Methods Find Exp Clin Pharmacol 2006;28: 177-83; Mackie and Stella, AAPS J 2006;8:E298-306; Pavlopoulos, Curr Pharm Des 200ό;12(14):1751-69.).
[00457] In certain embodiments, the cannabinoid agonist useful for the present invention may be selected from the group consisting of dexanabinol (HU211), BAY 38-7271 , Naphthalen- 1 -yl-(4-pentyloxynaphthalen- 1 -yl)methanone, THC (delta-9-tetrahydrocannabinol), nabilone, dronabinol, cannabidiol, cannabinol, cannabichromene, cannabigerol, cannabigerol, anandamide, (R)- methanandamide, 2-arachidonoylglycerol, HU210, desacetyllevonantxadol, CP55940, CP55244, URB602, or WIN55212-2 and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof.
[00458] In certain embodiments, the cannabinoid agonist useful for the present invention may be selected from the group consisting of 9-THC propyl analog, endocannabinoids, cannabinoid terpenoids, cannabinoid flavonoids, inhibitors of cannabinoid agonist metabolism, inhibitors of monoacylglycerol lipase, cannabidiol propyl analogues, cannabichromene propyl analogues, THC-like ABC tricyclic cannabinoid analogues, synthetic AC bicyclic cannabinoid analogues, synthetic ACD tricyclic cannabinoid analogues, aminoalkylindole compounds or analogs of 2-Arylimino-5,6-dihydro-4H-l,3-thiazines and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof.
[00459] In certain embodiments, the amount of the cannabinoid agonist used by the subject may be from about 10 ng to about 2000 mg, even up to about 2000 mg. More preferably, the amount of the cannabinoid agonist is from about 10 ng to about 1500 mg, even more preferably from about 0.1 mg to about 1000 mg, and most preferably, from about 0.1 mg to about 700 mg.
[00460] In some preferred embodiments, the cannabinoid agonist may be selected from compounds disclosed in U.S. Patent No. 7,217,732, 7,214,716, 7,169,942, 7,109,216, 7,091,216, 7,057,051, 6,995,184, 6,972,295, 6,943,266, 6,903,137, 6,864,291, 6,864,285, 6,525,087, 6,524,805, 6,509,367, 6,284,788, 5,948,777, 5,939,429, and 5,605,906, and in U.S. Patent Application No. 20070167514, 20070123505, ' 20070105914, 20070099947, 20070088058, 20070088025, 20070087390, 20070060638, 20070032517, 20070027144, 20060293299, 20060241 165, 20060172019, 20060106071, 20060089356, 20060079557, 20060074086, 20050272763, 20050267161, 20050245554, 20050239828, 20050239133, 20050234061, 20050203112, 20050182103, 20050165118, 20050154202, 20050137173, 20050101542, 20050096379, 20050065189, 20050054679, 20050026986, 20050009902, 20040266861, 20040266841, 20040248956, 20040242593, 20040235854, 20040229928, 20040229850, 20040171613, 20040106614, 20040058820, 20040044051, 20040034090, 20040018151, 20030175822, 20030138508, 20030114495, 20020173528, 20020128302, 20020077322, and 20010034344, and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof. All of the above patents and patent applications are hereby incorporated by reference in their entirety.
[00461] For purposes of the present invention, the term "cannabinoid agonist" shall include combinations of more than one cannabinoid agonist, and also include the unsalified agonist, mixed agonist-antagonists, partial agonists, pharmaceutically acceptable salts thereof, stereoisomers thereof, ethers and esters thereof, and mixtures thereof.
[00462] Notwithstanding the above definitions of "cannabinoid agonist", for the purposes of the present invention, 1) drugs that enhance the effect of cannabinoid agonists by inhibiting their metabolism or reuptake (for example, anandamide amidase inhibitors) are considered to be cannabinoid agonists; 2) drugs that induce anandamide amidase inhibitor metabolism or induce CB], CB2 and non-CBi/non-CB2 cannabinoid agonist metabolism or enhance reuptake will be considered cannabinoid antagonists; 3) inverse cannabinoid agonists will be considered cannabinoid antagonists.
[00463] A number of assays are available to determine whether a drug is a cannabinoid agonist, using in vivo and in vitro bioassay systems (Howlett et al., MoI Pharmacol, 1988; International Union of Pharmacology [IUPHAR], http://www.iuphar.org/index.html; Subcommittees on Cannabinoid Receptors The International Committee of Pharmacology Committee on Receptor Nomenclature and Classification [NC-IUPHAR], http ://www. iuphar . org/nciuphar .html) .
[00464] Notwithstanding the above definitions, for the purposes of the present invention, drugs that enhance the effect of cannabinoid agonists by inhibiting their metabolism or reuptake (for example, anandamide amidase inhibitors or inhibitors of monoacylglycerol lipase, which catalyzes 2-arachidonoylglycerol hydrolysis ) are also considered to be cannabinoid agonists.
[00465] The term "cannabinoid agonist" means a substance that binds to one or more cannabinoid receptor to exert an agonist or partial agonist effect.
[00466] The term "cannabinoid receptor" means a molecule that causes a specific physiologic, pathophysiologic or pharmacologic effect after binding to CBi, CB2, non-CBj/CB2 cannabinoid sites, TRPVi receptors, as well as other G protein-coupled receptors (GPCRs) that form part of the endocannabinoid system (Wiley and Martin, Chemistry Physics of Lipids, 2002; Begg et al., Pharmacol Ther, 2005; Howlett et al., Neuropharmacol, 2004; Pertwee, AAPS Journal, 2005; International Union of Pharmacology (IUPHAR) Receptor Database; Howlett et al., MoI Pharmacol, 1988; International Union of Pharmacology [IUPHAR], http://www.iuphar.org/index.html; Subcommittees on Cannabinoid Receptors The International Committee of Pharmacology Committee on Receptor Nomenclature and Classification [NC-IUPHAR], http://www.iuphar.org/nciuphar.html).
[00467] The term "opioid receptor" includes mu (μ), delta (δ), kappa (K) and/or nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptors, their subtypes and splice variants such as μ1 ; μ2, δi, 62, Ki, K2 and K3, etc, regardless of whether they also bind to or influence other receptor systems (e.g., norepinephrine reuptake inhibition, serotonin reuptake inhibition, NMDA receptor antagonism).
[00468] Opioid antagonists are known or readily determined by individuals who practice the art. Preferably, the opioid antagonists useful for the present invention may be selected from the group consisting of naltrexone, methylnaltrexone, nalbuphine, naloxone, nalmefene, cyclazocine, cyclorphan, oxilorphan nalorphine, nalorphine dinicotinate, nalmefene, nadide and levallorphan.
[00469] Abusable drugs of the invention include stimulants, CNS-stimulants, psychostimulants, alkylxanthine, and anorectics, including, without limitation, compounds selected from the group comprising adrafanil, alkyxanthine derivatives, almitrine, amfetaminil, aminophylline, amiphenazole, ammonium camphocarbonate, amphetamine, bamifylline, benzfetamine, brolamfetamine, caffeine, cathine [(+)-norpseudoephedrine], cathinone, celastrin, chloφhentermine, clonobenzorex, cropropamide, crotetamide, deanol, dextroamphetamine, diethylaminoethanol, diethylpropion, dimfline, doxapram, doxofylline, diprophylline, dyphylline, etamivan, etofylline, enprophylline, etamiphylline, methylphenidate, dexmethylphenidate, fencamfamin, fenetylline, fenozolone, fenproporex, lisdexamfetamine, lisdexamfetamine dimesylate, lisofylline, lobeline, mazindol, mefenorex, mepixanox, methamphetamine, methylenedioxymethamphetamine, modafinil, nicotine, nikethamide, oxtriphylline, pemoline, pentetrazol, phedimetrazine, phenmetrazine, phentermine, pentoxifylline, phenylpropanolamine, pipradrol, prethcamide, prolintane, propylhexedrine, propentofylline, pentifylline, pseudoephedrine, pyridophylline, proxyphylline, sibutramine, tenamfetamine (methylenedioxeamphetamine), theophylline, theobromine, and their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof.
[00470] The present invention anticipates the use of more than one abusable drug in some embodiments, given in the same formulation or in a different formulation, for use to treat, prevent or ameliorate the same disease or a different disease.
[00471] In certain preferred embodiments of the present invention, the invention allows for the use of lower doses of abusable drug by virtue of the inclusion or co-administration of an additional drug for the prevention or treatment of the sam medical condition. By using lower amounts of either or both drugs, the side effects associated with treatment in humans are reduced.
[00472] The term "abusable drug" means an the abusable drug in unsalified form, a pharmaceutically acceptable salt, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixture thereof.
[00473] The singular forms "a", "an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "ADER" includes an ADER compound as well as a mixture of two or more different ADER compounds, reference to "cannabinoid agonists" includes a cannabinoid agonist as well as two or more different cannabinoid agonists in combination, and the like.
[00474] As used herein, the term "analgesic" includes pharmacologic agents intended for or effective in the prevention and/or treatment of pain.
[00475] In some embodiments, in addition to preventing or treating pain, analgesics provide salutary effects on signs and symptoms associated with pain. For example, analgesics, in addition to relieving pain in patients with osteoarthritis, relieve stiffness, improve physical function, sleep and quality of life. For example, analgesics, in addition to relieving pain in patients with neuropathic pain, reduce disability.
[00476] As used herein, the term "pain" includes: (i) peripheral neuropathic pain, e.g., acute and chronic inflammatory demeyelinating polyradiculopathy, alcoholic polyneuropathy, chemotherapy-induced polyneuropathy, complex regional pain syndrome (CRPS) Type I and Type II, entrapment neuropathies (e.g., carpal tunnel syndrome), HIV sensory neuropathy, iatrogenic neuralgias (e.g., postthoracotomy pain, postmastectomy pain), idiopathic sensory neuropathy, painful diabetic neuropathy, phantom limb pain, postherpetic neuralgia, trigeminal neuralgia, radiculopathy (e.g., cervical thoracic, lumbosacral), sciatica, acute herpes zoster pain, temporomandibular joint ■disorder pain and postradiation plexopathy; and (ii) central neuropathic pain, e.g., compressive myelopathy from spinal stenosis, HIV myelopathy, multiple sclerosis pain, Parkinson's disease pain, postischemic myelopathy, post postradiation myelopathy, poststroke pain, posttraumatic spinal cord injury and syringomyelia; and (iii) cancer associated neuropathic pain, e.g., chemotherapy induced polyneuropathy, neuropathy secondary to tumor infiltration or nerve compression, phantom breast pain, postmastectomy pain, postradiation plexopathy and myelopathy; (iv) chronic pain, e.g., back pain, rheumatoid arthritis, osteoarthritis, inflammatory pain, non-inflammatory pain, myofascial pain, fibromyalgia, cancer pain, visceral pain, somatic pain, pelvic pain, musculoskeletal pain, post-traumatic pain, bone pain and idiopathic pain; (v) acute pain, e.g, acute postsurgical pain (including laparoscopic, laparatomy, gynecologic, urologic, cardiothoracic, arthroscopic, gastrointestinal, neurologic, orthopedic, oncologic, maxillofacial, ophthalmic, otolaryngologic, soft tissue, plastic, cosmetic, vascular and podiatric surgery, including abdominal surgery, abdominoplasty, adenoidectomy, amputation, angioplasty, appendectomy, arthrodesis, arthroplasty, arthroscopy, bilateral cingulotomy, biopsy, brain surgery, breast biopsy, cauterization, cesarean section, cholecystectomy, circumcision, commissurotomy, cordotomy, corneal transplantation, cricothoracotomy, discectomy, diverticulectomy, episiotomy, endarterectomy, endoscopic thoracic sympathectomy, foreskin restoration, fistulotomy, frenectomy, frontalis lift, fundectomy, gastrectomy, grafting, heart transplantation, hemicorporectomy, hemorrhoidectomy, hepatectomy, hernia repair, hypnosurgery, hysterectomy, kidney transplantation, laminectomy, laparoscopy, laparotomy, laryngectomy, lithotripsy, lobotomy, lumpectomy, lung transplantation, mammectomy, mamrnoplasty, mastectomy, mastoidectomy, mentoplasty, myotomy, mryingotomy, nephrectomy, nissen fundoplication, oophorectomy, orchidectomy, parathyroidectomy, penectomy, phalloplasty, pneumotomy, pneumonectomy, prostatectomy, psychosurgery, radiosurgery, ritidoplasty, rotationplasty, sigmoidostomy, sphincterotomy, splenectomy, stapedectomy, thoracotomy, thrombectomy, thymectomy, thyroidectomy, tonsillectomy, tracheotomy, tracheostomy, tubal ligation, ulnar collateral ligament reconstruction, ureterosigmoidostomy, vaginectomy, vasectomy, vulvectomy; renal colic; incisional pain; inflammatory incisional pain; nociceptive incisional pain; acute neuropathic incisional pain following surgery), renal colic, trauma, acute back pain, burn pain, burn dressing change pain, migraine pain, tension headache pain, acute musculoskeletal pain, acute exacerbation or flare of chronic back pain, acute exacerbation or flare of osteoarthritis, acute exacerbation or flare of chronic pain, breakthrough chronic non-cancer pain, breakthrough cancer pain, acute exacerbation or flare of fibromylagia, acute exacerbation or flare of rheumatoid arthritis, acute exacerbation or flare of myofacsial pain, acute exacerbation or flare of chronic idiopathic pain, acute exacerbation or flare of neuropathic pain, procedure related pain (e.g., arthroscopy, laparoscopy, endoscopy, intubation, bone marrow biopsy, soft tissue biopsy, catheterization), and other self-limiting pain states.
[00477] As used herein, the term "acute pain" refers to self-limiting pain that subsides over time and usually lasting less that about 30 days and more preferably lasting less than about 21 days. Acute pain does not include chronic conditions such as chronic neuropathy, chronic neuropathic pain and chronic cancer and non-cancer pain.
[00478] As used herein, "neuropathic pain" is pain initiated or caused by a primary lesion or dysfunction of the nervous system and includes (i) peripheral neuropathic pain and (ii) central neuropathic pain.
[00479] As used herein, the term "chronic pain" includes all non-neuropathic pain lasting more than 30 days, including inflammatory pain, noninflammatory pain, muscle pain, joint pain, fascia pain, visceral pain, bone pain and idiopathic pain.
[00480] As used herein, "Attention-Deficit/Hyperactivity Disorder" or
"ADHD" is disorder as described in the Diagnostic and Statistical Manual of Mental Disorders (DSM), 1994, as revised in 2000 (DSM-IV-TR). ADHD is generally considered to be a developmental disorder, largely or entirely neurological in nature effecting 3-5 percent of the population characterized by a persistent pattern of inattention and/or hyperactivity-impulsivity, including hyperactivity, forgetfulness, poor impulse control, inattention, difficulty getting work done, procrastination, or organization problems and distractibility.
[00481] As used herein, "neurologic disorders" are disorders that affect the central nervous system (brain and spinal cord), the peripheral nervous system (peripheral nerves - cranial nerves included), or the autonomic nervous system (parts of which are located in both central and peripheral nervous system). See Adams & Victor's Principles of Neurology (McGraw-Hill Professional; 7 edition, 2000); Merritt's Textbook of Neurology (9th ed. Edited by Lewis P. Rowland. Baltimore: Williams and Wilkins, 1995); and Guide to Clinical Neurology (Mohr and Gautier, eds, New York, Churchill Livingstone, 1995).
[00482] The term "psychiatric disorders" and "mental illness" are used interchangeably. A mental illness is an abnormal mental condition or disorder expressing symptoms that cause significant distress and/or dysfunction. This can involve cognitive, emotional, behavioral and interpersonal impairments. As used herein, psychiatric disorders are disorders described in the Diagnostic and Statistical Manual of Mental Disorders (DSM), 1994, as revised in 2000 (DSM-IV-TR).
[00483] The term "analgesic effectiveness" is defined for purposes of the present invention as a satisfactory prevention, reduction in or elimination of pain, along with a tolerable level of side effects, as determined by the human patient.
[00484] "Therapeutically effective amount", "therapeutic effectiveness" or
"therapeutically-effective" refers to the amount of an active agent sufficient to induce a desired biological result. That result may be alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
[00485] The term "effective amount" means the quantity of a compound according to the invention necessary to prevent, to cure, or at least partially arrest a sign or symptom for which the compound (e.g., abusable drug) has been prescribed to a subject.
[00486] The term "abuse resistant" and "abuse deterrent" are used interchangeably and include pharmaceutical compositions and methods to resist intentional, unintentional or accidental physical, mechanical, chemical or thermal manipulation or tampering of the dosage form (e.g., crushing, shearing, grinding, pulverizing, chewing, dissolving, melting, needle aspiration, syringe aspiration, syringe injection, solvent extraction, inhalation, insufflation, extraction by mechanical, thermal and chemical means, and/or filtration). The term "abuse resistant" and "abuse deterrent" also includes pharmaceutical compositions and methods to resist intentional, unintentional or accidental use or misuse of the dosage form: (i) in quantities or by methods and routes of administration that do not conform to standard medical practice; (ii) outside the scope of specific instructions for use provided by a qualified medical professional; (iii) outside the supervision of a qualified medical professional; (iv) outside the approved instructions on proper use provided by the drug's legal manufacturer; (v) in unapproved dosage forms; (vi) for compulsive use; (vii) through acquisition by manipulation of the medical system; (viii) for medically unapproved or unintended mood altering purposes.
[00487] "Drug", "drug substance", "substance", "therapeutic agent",
"pharmacological agent", "pharmaceutical agent", and "active agent" are used interchangeably and are intended to have their broadest interpretation as to any therapeutically active substance which is delivered to a living organism to produce a desired, usually beneficial effect. In general, this includes therapeutic agents in all of the major therapeutic areas.
[00488] The term "subject" for purposes of treatment is used interchangeably with "patient", "male", "female", and includes any human subject.
[00489] As used herein, "bioequivalent" and "bioequivalence" means that the
90% Confidence Interval (CI) of the relative mean Qrax, AUC(o-t) and AUQo-∞) of the drug under test and reference conditions (e.g., generic vs. brand name, or fed versus fasted, or with and without concurrent alcohol) is within 80% to 125%, when tested in accordance with U.S. FDA guidelines (see "Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products-General Considerations", Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research, July 2002 and "Guidance for Industry: Food-Effect Bioavailability and Fed Bioequivalence Studies: Study Design, Data Analysis and Labeling", Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research, October, 2001, which are hereby incorporated by reference).
[00490] "Pharmaceutically or therapeutically acceptable excipient or carrier" or
"excipient" refers to a substance which does not interfere with the effectiveness or the biological activity of the active ingredients and which is not toxic to the subject. In some preferred embodiments of the present invention, pharmaceutically or therapeutically acceptable excipients or carriers may play a role in imparting or optimizing the rate and extent of absorption of the abusable drug or additional drugs in the pharmaceutical composition. In some preferred embodiments of the present invention, pharmaceutically or therapeutically acceptable excipients or carriers may play a role in stabilizing the abusable drug or additional drugs in the pharmaceutical composition.
[00491] In certain preferred embodiments of the present invention, the dosage form may include, in addition to abusable drug or a pharmaceutically acceptable salt thereof and ADER5 other abuse deterrent or abuse resistant substances, process or technologies known in the art, including one or more aversive agents. All kinds of aversive agents are contemplated, including, without limitation, antagonists of abusable drugs, laxatives, cutaneous vasodilators, headache producing agents, emetics, emetogenic compound, nausea producing compounds, bittering agents, drugs that cause burning on irritation when in contact with tissue or mucous membranes (e.g., naso- mucosal irritants, oro-mucosal irritants, respiratory irritants), tissue irritants, gastrointestinal irritants, drugs that precipitate withdrawal effects, tissue dyes, lakes and colorants, beverage dyes, lakes and colorants, non-tissue staining beverage dyes, lakes and colorants (i.e, that do not stain or discolor the skin upon ingestion), fecal discolorants, urine discolorants, malodorous agents, opioid antagonists, benzodiazepine antagonists (e.g., flumazenil), cannabinoid antagonists and pharmacologic antagonists to co-abused drugs not contained in the dosage form. Such aversive agents may be in the dosage form in a releasable, partially releasable or a non-releasable form (i.e., sequestered), the latter being released on tampering the dosage form (e.g., mechanical, thermal, chemical, solvent tampering, ingestion in ways not recommended, and the like). Further, in some embodiments, such aversive agents may be in the dosage form in an amount that does not produce an aversive effect or aversion in any, many or substantially all patients when taken in accordance with the prescribing information or the manufacturer's instructions (for example, in small quantities), but which produce an aversive effect when taken in excess (e.g., higher dose or more frequently).
[00492] In some embodiments, one or more aversive agents may be added to the formulation in an aversive agent amount of less than about 80% by weight, preferably less than about 60% by weight, more preferably less than about 40% by weight of the dosage form, even more preferably less than about 20% by weight of the dosage form, and most preferably less than about 10 by weight of the dosage form (e.g., 0.000000000000001% to 1%, or 0.000000001% to 3%, or 0.0001% to 10%, or 0.001% to 5%, or 1% to 10%, or 0.001% to 2%, or 1% or 10%, or 2% to 7%) depending on the particular aversive agent used.
[00493] In some embodiments, the aversive agent in the dosage form may be about 0.00000000001 mg to about 2000 mg, or about 0.0000001 mg to about 1500 mg, or about 0.000001 mg to about 1000 mg, or about 0.0001 mg to about 1000 mg, or about 0.001 mg to about 1000 mg, or about 0.01 mg to about 1000 mg, or about 0.1 rag to about 1500 mg, or 1 mg to about 800 mg, or about 1 mg to about 500 mg, or about 1 mg to about 300 mg, or about 1 mg to about 150 mg, or about 5 mg to about 400 mg, or about 5 mg to about 200 mg, or about 0.00000000001 mg to about 200 mg, or about 0.00000000001 mg to about 100 mg, or about 0.00000000001 mg to about 50 mg, or about 0.0000001 mg to about 200 mg, or about 0.0000001 mg to about 100 mg, or about 0.00001 mg to about 400 mg, or about 0.0001 mg to about 300 mg.
[00494] As described above, the present invention can include one or more aversive agents, selected from the group including, without limitation antagonists of abusable drugs, laxatives, cutaneous vasodilators headache producing agents, emetics, emetogenic compound, nausea producing compounds, bittering agents, drugs that cause burning on irritation when in contact with tissue or mucous membranes (e.g., naso-mucosal irritants, oro- mucosal irritants, respiratory irritants), tissue irritants, gastrointestinal irritants, drugs that precipitate withdrawal effects, tissue dyes, lakes and colorants, beverage dyes, lakes and colorants, non-tissue staining beverage dyes, lakes and colorants, fecal discolorants, urine discolorants, malodorous agents, opioid antagonists, benzodiazepine antagonists, cannabinoid antagonists, and pharmacologic antagonists to co-abused drugs not contained in the dosage form. Preferably, the aversive agent is a pharmaceutically acceptable agent that produces an aversive effect only when the dosage form of the invention containing the aversive agent is abused, for example, when taken in excess of medically approved doses, taken in excess of approved doses in the manufacturer's prescribing information, taken after tampering of the dosage form (e.g., mechanical, thermal, chemical, solvent tampering), ingestion in ways not medically recommended, administration by routes not approved for the dosage form (e.g., intranasal, inhalation, intravenous) or in a manner inconsistent with the manufacturer's prescribing information. ] In some embodiments, the amount of aversive agent in the dosage form of the present invention can be a fixed ratio in relation to the amount of abusable drug in the dosage form. By appropriately selecting the quantity of the aversive agent in the dosage form, aversive effects can be avoided under conditions of proper medical use (e.g., manufacturers prescribing directions). However, under some conditions of abuse, for example excessive intake of the dosage form of the invention, the quantity of aversive agent consumed will exceed the "no effect" or "minimum effect" threshold, thereby producing one or more aversive effects, for example, e.g., nausea, emesis, diarrhea, laxation, cutaneous vasodilation, headache, bitter taste, naso-mucosal irritation, oro- mucosal irritation, precipitation of abstinence from the abusable drug of the dosage form, precipitation of abstinence from a co-abused drug which is not part of the dosage form, reduction of the pleasurable, mood altering, rewarding, reinforcing, stimulant, depressant or other psychic and physiologic effects of the abusable drug or a co-abused drug, etc.).
[00496] In some embodiments, the "no effect" or "minimum effect" threshold amount of aversive agent can be exceeded when the dosage form of the invention is taken in excess of the manufacturer's recommendation by a factor of about 1.5, or about 2, or about 2.5, or about 3, or about 4, or about 5, or about 6, or about 7, or about 8, or about 10, or more than 10. In some embodiments, the production of an aversive effect can reduce or stop further abuse of the dosage form, thereby reducing the harm or toxicity of the drug in the subject who is tampering, misusing or abusing the dosage form, e.g., addicts, drug abusers and recreational drug users.
[00497] Various bittering agents can be employed including, for example and without limitation, T2R or TAS2R receptor agonists, phenylthiourea (phenylthiocarbamide), natural, artificial and synthetic flavor oils and flavoring aromatics and/or oils, oleoresins and extracts derived from plants, leaves, flowers, fruits, and so forth, and combinations thereof. Nonlimiting representative flavor oils include spearmint oil, peppermint oil, eucalyptus oil, oil of nutmeg, allspice, mace, oil of bitter almonds, menthol and the like. Also useful bittering agents are artificial, natural and synthetic fruit flavors such as citrus oils including lemon, orange, lime, grapefruit, and fruit essences and so forth. Additional bittering agents include sucrose derivatives (e.g., sucrose octaacetate), chlorosucrose derivatives, quinine and quinine salts, quinidine and quindine salts and the like. The preferred bittering agent for use in the present invention is denatonium, denatonium benzoate and denatonium saccharide. A dosage form including a bittering agent preferably discourages improper usage of the tampered dosage form by imparting a disagreeable taste to the tampered dosage form.
[00498] In some embodiments, the aversive agent in the dosage form may be denatonium, denatonium saccharide or denatonium benzoate, in a quantity expressed as mg of denatonium, of about 0.00000001 nag to about 100 mg, or about 0.000001 mg to about 100 mg, or about 0.0001 mg to about 100 mg, or about 0.0001 mg to about 20 mg, or about 0.0001 mg to about 10 mg, or about 0.0001 mg to about 5 mg, or about 0.0001 mg to about 2 mg, or about 0.0001 tng to about 1 mg, about 0.0001 mg to about 50 mg, or about 0.00000001 mg to about 50 mg, or about 0.00000001 mg to about 20 mg, or about 0.01 mg to about 20 mg, or about 0.01 mg to about 10 mg, or about 0.01 mg to about 5 mg, or about 0.01 mg to about 2 mg, or about 0.01 mg to about 1 mg, or about 0.01 mg to about 1 mg, or about 0. 01 mg to about 0.5 mg, or about 0.1 mg to about 20 mg, or about 0.1 mg to about 10 mg, or about 0.1 mg to about 7 mg, or about 0.1 mg to about 5 mg, or about 0.1 mg to about 3 mg, or about 0.1 mg to about 2 mg.
[00499] In some embodiments, the aversive agent in the dosage form may be quinine or a pharmaceutically acceptable salt of quinine, in a quantity expressed as mg of quinine, of about 0.00001 mg to about 300 mg, or about 0.00001 mg to about 200 mg, or about 0.00001 mg to about 100 mg, or about 0.00001 mg to about 75 mg, or about 0.00001 mg to about 50 mg, or about 0.00001 mg to about 25 mg, or about 0.00001 mg to about 20 mg, or about 0.00001 mg to about 10 mg, or about 0.00001 mg to about 5 mg, or about 0.00001 mg to about 2.5 mg, or about 0.00001 mg to about 1 mg, or about 0.001 mg to about 300 mg, or about 0.001 mg to about 200 mg, or about 0.001 mg to about 100 mg, or about 0.001 mg to about 75 mg, or about 0.001 mg to about 50 mg,.or about 0.001 mg to about 25 mg, or about 0.001 mg to about 20 mg, or about 0.001 mg to about 10 mg, or about 0.001 mg to about 5 mg, or about 0.001 mg to about 2.5 mg, or about 0.001 mg to about 1 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 100 mg, or about 1 mg to about 75 mg, or about 1 mg to about 50 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 1 mg to about 2.5 mg.
[00500] Various emetic agents can be employed including, for example and without limitation, zinc and pharmaceutically acceptable salts thereof (e.g., zinc oxide, zinc gluconate, zinc acetate, zinc sulfate, zinc carbonate), dopamine agonists, apomorphine, ipecac, ipecacuanha, emetine, emetine (methylcephaeline), cephaeline, psychotrine, O-methylpsychotrine, ammonium chloride, potassium chloride, magnesium sulfate, ferrous gluconate, ferrous sulfate, aloin, algarot or antimonious oxychloride, antimony trichloride, folate, folic acid, niacin (niacin) and nicotinamide. In some embodiments, the aversive agent in the dosage form may be zinc in the form of elemental zinc or a pharmaceutically acceptable salt of zinc, in a quantity expressed as mg of elemental zinc, of about 1 mg to about 400 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 150 mg, or about 1 mg to about 100 mg, or about 1 mg to about 90 mg, or about 1 mg to about 80 mg, or about 1 mg to about 70 mg, or about 1 mg to about 60 mg, or about 1 mg to about 50 mg, or about 1 mg to about 45 mg, or about 1 mg to about 40 mg, or about 1 mg to about 40 mg, or about 1 mg to about 35 mg, or about 1 mg to about 30 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 5 mg to about 400 mg, or about 5 mg to about 300 mg, or about 5 mg to about 200 mg, or about 5 mg to about 150 mg, or about 5 mg to about 100 mg, or about 10 mg to about 150 mg, or about 10 mg to about 100 mg, or about 5 mg to about 80 mg, or about 5 mg to about 60 mg, or about 5 mg to about 50 mg, or about 5 mg to about 45 mg, or about 5 mg to about 40 mg, or about 5 mg to about 40 mg, or about 5 mg to about 35 mg, or about 5 mg to about 30 mg, or about 5 mg to about 25 mg, or about 5 mg to about 20 mg, or about 5 mg to about 10 mg, or about 10 mg to about 90 mg, or about 10 mg to about 80 mg, or about 10 mg to about 60 mg, or about 10 mg to about 50 mg, or about 10 mg to about 45 mg, or about 10 mg to about 40 mg, or about 10 mg to about 40 mg, or about 10 mg to about 35 mg, or about 10 mg to about 30 mg, or about 10 mg to about 25 mg, or about 10 mg to about 20 mg, or about 20 mg to about 100 mg, or about 20 mg to about 90 mg, or about 20 mg to about 80 mg, or about 20 mg to about 60 mg, or about 20 mg to about 50 mg, or about 20 mg to about 45 mg, or about 20 mg to about 40 mg, or about 20 mg to about 35 mg, or about 20 mg to about 30 mg, or about 15 mg to about 50 mg, or about 15 mg to about 40 mg, or about 15 mg to about 35 mg, or a quantity sufficient to be produce an aversive effect vasodilation when abused but not under conditions of medically appropriate use.
[00502] Various irritants can be employed including, for example and without limitation transient receptor potential vanilloid 1 (TRPVl or VRl) agonists (including resiniferanoids, capsaicinoids, phorboid vanilloids, and terpenoid 1,4-unsaturated dialdehydes, capsaicin, capsaicin analogs and derivatives, resiniferatoxin, olvanil, pipeline, zingerone, anandamide, 12- and 15-(S)- hydroperoxy-eicosatetraenoic acids, 5 and 15-(S)-hydroxyeicosatetraenoic acids, phorbol 12-phenylacetate 13-acetate 20-homovanillate, 2 phorbol 12,13- didecanoate 20-homovanillate, leukotriene B(4), tinyatoxin, heptanoylisobutylamide, N-(3 -acyloxy-2-benzylpropy 1 )-N'- dihydroxytetrahydrobenzazepine, tetrahydroisoquinoline thiourea analogs, heptanoyl guaiacylamide, other isobutylamides or guaiacylamides, dihydrocapsaicin, homovanillyl octylester and nonanoyl vanillylamide), acids such as acids with one or more carboxyl moieties (e.g., formic acid, acetic acid, propionic acidy, butyric acid, valeric acid, caproic acid, caprillic acid, capric acid, oxalic acid, malonic acid, succicnic acid, glutaric acid, adipic acid, maleic acid, fumaric acid, and citric acid), sodium lauryl sulfate, poloxamer, sorbitan monoesters, glyceryl monooleates, niacin, mustard, allyl isothiocyaanate and p-hydroxybenzyl isothiocyanate, acetylsalicylic acid.
[00503] Various cutaneous vasodilators can be employed including, for example and without limitation, niacin acid, nicotinuric acid, beta- hydroxybutyrate and nicotinic receptor (e.g., HM74A or GPRl 09A) agonists.
[00504] In some embodiments, the aversive agent in the dosage form may be niacin, in a quantity of about 1 nig to about 400 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 150 mg, or about 1 mg to about 100 mg, or about 1 mg to about 90 mg, or about 1 mg to about 80 mg, or about 1 mg to about 70 mg, or about 1 mg to about 60 mg, or about 1 mg to about 50 mg, or about 1 mg to about 45 mg, or about 1 mg to about 40 mg, or about 1 mg to about 40 mg, or about 1 mg to about 35 mg, or about 1 mg to about 30 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 5 mg to about 400 mg, or about 5 mg to about 300 mg, or about 5 mg to about 200 mg, or about 5 mg to about 150 mg, or about 5 mg to about 100 mg, or about 10 mg to about 150 mg, or about 10 mg to about 100 mg, or about 5 mg to about 80 mg, or about 5 mg to about 60 mg, or about 5 mg to about 50 mg, or about 5 mg to about 45 mg, or about 5 mg to about 40 mg, or about 5 mg to about 40 mg, or about 5 mg to about 35 mg, or about 5 mg to about 30 mg, or about 5 mg to about 25 mg, or about 5 mg to about 20 mg, or about 5 mg to about 10 mg, or or about 10 mg to about 90 mg, or about 10 mg to about 80 mg, or about 10 mg to about 60 mg, or about 10 mg to about 50 mg, or about 10 mg to about 45 rag, or about 10 mg to about 40 mg, or about 10 mg to about 40 mg, or about 10 mg to about 35 mg, or about 10 mg to about 30 mg, or about 10 mg to about 25 mg, or about 10 mg to about 20 mg. or about 20 mg to about 100 mg, or about 20 mg to about 90 mg, or about 20 mg to about 80 mg, or about 20 mg to about 60 mg, or about 20 mg to about 50 mg, or about 20 mg to about 45 mg, or about 20 mg to about 40 mg, or about 20 mg to about 35 mg, or about 20 mg to about 30 mg, or about 15 mg to about 50 mg, or about 15 mg to about 40 mg, or about 15 mg to about 35 mg, or a quantity sufficient to be produce an aversive effect when abused but not under conditions of medically appropriate use. 05] Various tissue dyes, lakes and colorants, beverage dyes, lakes and colorants, non-tissue staining beverage dyes, lakes and colorants, fecal discolorants, urine discolorants can be employed including, for example and without limitation, Curcumin, Riboflavin, Tartrazine, Quinoline yellow, Sunset yellow FCF, Carmine, Carmoisine, Amaranth, Ponceau 4R, Erythrosine, Allura red AC, Patent blue V, Indigo carmine, Brilliant blue FCF, Chlorophylls, Copper complexes of chlorophylls and chlorophyllins, Green S, i Caramel, Brilliant black BN, Vegetable carbon, Carotenoids, Alpha-, beta-, gamma-carotene, Capsanthin, Capsorubin, Lycopene, Beta-apo-8' carotenal, Ethyl ester of beta-apo-8' carotenoic acid, Xanthophylls, Lutein, Canthaxanthin, Beetroot red, Anthocyanins, Cyanidin, Delphidin, Malvidin, Pelargonidin, Peonidin, Petunidin, Calcium carbonate, Titanium dioxide, Iron oxides and hydroxides, Aluminum, Brilliant blue FCF, Indigotine, Alphazurine FG, Indanthrene blue, Fast green FCF, Alizarin cyanine green F, Quinizarine green SS, Pyranine concentrated, Orange II, Dibromofluorescein, Diiodofluorescein, Erythrosine yellowish Na, Erythrosine, Ponceau SX, Lithol rubin B, Lithol rubin B Ca, Toney red, Tetrabromofluorescein, Eosine, Tetrachlorotetrabromofluorescein, Phloxine B, Helindone pink CN, Brilliant lake red R, Acid fuchsine, Lake bordeaux B, Flaming red, Alba red, Allura red AC, Allura Red AC, Alizurol purple SS, Tartrazine, Sunset yellow, FCF, Fluorescein, Naphthol yellow S, Uranine, Quinoline yellow WS, Quinoline yellow SS, Brilliant blue FCF, Indigotine, Alphazurine FG, Alizurol purple SS, Sunset yellow FCF, Alumina, Aluminum powder, Annatto extract, Beta- carotene, Bismuth oxychloride, Bronze powder, Calcium carbonate, Canthaxanthin, Caramel, Chromium-cobalt-aluminum oxide, Chromium hydroxide green, Chromium oxide green, Cochineal extract, carmine, Copper powder, Dihydroxyacetone, Ferric ammonium citrate, Ferric ammonium ferrocyanide, Ferric ferrocyanide, Guanine, Iron oxides synthetic, Logwood extract, Mica, Potassium sodium copper chlorophyllin, Pyrogallol, Pyrophyllite, Talc, Titanium dioxide, Zinc oxide, FD&C blue #1, FD&C blue #2, D&C blue #4, D&C blue #9, FD&C green #3, D&C green #5, D&C green #6, D&C green #8, D&C orange #4, D&C orange #5, D&C orange #10, D&C orange #11, FD&C red #3, FD&C red #4, D&C red #6, D&C red #7, D&C red #17, D&C red #21, D&C red #22, D&C red #27, D&C red #28, D&C red #30, D&C red #31, D&C red #33, D&C red #34, D&C red #36, D&C red #39, FD&C red #40, FD&C red #40 lake, D&C violet #2, FD&C yellow #5, FD&C yellow #6, D&C yellow #7, Ext. D&C yellow #7, D&C yellow #8, D&C yellow #10, D&C yellow #11, FD&C lakes, D&C lakes, Ext. D&C lakes, FD&C blue #1 lake, FD&C blue #2 lake, D&C blue #4 lake, FD&C green #3 lake, D&C green #5 lake, D&C green #6 lake, D&C orange #4 lake, D&C orange #5 lake, D&C orange #10 lake, D&C orange #11 lake, FD&C red #4 lake, D&C red #6 lake, D&C red #7 lake, D&C red #17 lake, D&C red #21 lake, D&C red #22 lake, D&C red #27 lake, D&C red #28 lake, D&C red #30 lake, D&C red #31 lake, D&C red #33 lake, D&C red #34 lake, D&C red #36 lake, D&C violet #2 lake, FD&C yellow #5 lake, FD&C yellow #6 lake, D&C yellow #7 lake, Ext. D&C yellow #7 lake, D&C yellow #8 lake, D&C yellow #10 lake, Turmeric, Lactoflavin, Cochineal, carminic acid, Indigotine, Magnesium chlorophyll, Brilliant green BS, Black PN, Carbo medicinalis vegetabilis, Paprika oleoresin, Paprika oleoresin, Betanin, Beta-carotene, indigo carmine, iron oxides, sunset yellow FCF, titanium dioxide, ElOO, ElOl, E102, E104, EIlO, E120, E122, E123, E124, E127, E129, E131, E132, E133, E140, E141, E142, E150, E151, E153, E160, E161, E162, E163, E170, E171, El 72, El 73 and phenazopyridine.
[00506J As used herein, "dyes", "lakes", "colorants" and "discolorants" are used interchangeably and refer to one or more pharmaceutically acceptable dyes, lakes or colorants which may be: (i) tissue staining; (ii) non-tissue staining; (iii) beverage staining; (iv) urine discolorant; and/or (v) fecal discolorant.
[00507] Various laxatives can be employed including, for example and without limitation, Bis(p-hydroxyphenyl)pyridyl-2-rnethane, Bisacodyl, bisoxatin, anthraquinone, anthraquinone analogs and derivatives (e.g., buckthorn, casanthranol, cascara, hydroxy anthracene, glucofrangulin ), dantron, danthron, docusate (e.g., docusate sodium, docusate calcium, docusate potassium), gastrointestinal chloride channel activators (e.g., chloride channel subtype 2 activators), lubiprostone, magenesium salts (e.g., magnesium citrate, magnesium hydroxide, magnesium oxide), mannitol, oxyphenisatine, polyethylene glycol, poly(ethylene oxide) [PEO-1500], sodium phosphate, phenolphthalein, senna, senna constituents and derivatives (e.g., sennoside A, sennoside B) and sodium picosulfate.
[00508] In some embodiments, the aversive agent in the dosage form may be a laxative in the amount of about 0.001 mg to about 300 mg, or about 0.001 mg to about 200 mg, or about 0.001 mg to about 100 mg, or about 0.001 mg to about 75 mg, or about 0.001 mg to about 50 mg, or about 0.001 mg to about 25 mg, or about 0.001 mg to about 20 mg, or about 0.001 mg to about 10 mg, or about 0.001 mg to about 5 mg, or about 0.001 mg to about 2.5 mg, or about 0.001 mg to about 1 mg, or about 1 mg to about 300 mg, or about 1 mg to about 200 mg, or about 1 mg to about 100 mg, or about 1 mg to about 75 mg, or about 1 mg to about 50 mg, or about 1 mg to about 25 mg, or about 1 mg to about 20 mg, or about 1 mg to about 10 mg, or about 1 mg to about 5 mg, or about 1 mg to about 2.5 mg.
[00509] Aversive agents may include compounds found on the FDA EAFUS database (http://vm.cfsan.fda.gov/~dms/eafus.html); FDA Food Additives Status List (http://www.cfsan.fda.gov/~dms/opa-appa.html); FDAGRAS list and database; FDA Color Additive Status List
(http://www.cfsan.fda.gov/~dms/opa-appc.html); FDA Inactive Ingredients Database (http://www.accessdata.fda.gov/scripts/cder/iig/index.cfrn); Rowe, Sheskey and Owen, Handbook of Pharmaceutical Excipients, APhA Publications; 5th edition (2006); Goodman & Gilman's The Pharmacological Basis of Therapeutics (Brunton, Lazo and Parker, eds, 11th ed., McGraw Hill (2005); Remington: The Science and Practice of Pharmacy, 21st ed, Lippincott Williams & Wilkins (2005); Martindale: The Complete Drug Reference, 35th Edition, Pharmaceutical Press (2007); United States Pharmacopeia-National Formulary (USP-NF), (USP 30 - NF 25, 2007), the International Programme on Chemical Safety (http://www.inchem.org/) and Health Canada's List of Acceptable Non-medicinal Ingredients (http://www.hc-sc.gc.ca/dhp- mps/prodnatur/legislation/docs/nmi-imn_listl_e.html), all hereby incorporated by reference in their entirety.
[00510] It should be noted that the above mentioned aversive agents may, in some embodiments be used in the dosage form of the invention for purposes other than as aversive agents, or for both aversive and non-aversive purposes. Such non-aversive uses can include, without limitation, pharmaceutical purposes and pharmacologic purposes. For example, in some embodiments, the laxative agent may be used to counteract the constipating effects of the abusable dosage form of the invention. In some embodiments, zinc and pharmaceutically acceptable salts of zinc and niacin may be used for pharmaceutical purposes (e.g., pharmaceutical optimization, drug release and drug stability).
[00511] hi one preferred embodiment of the invention, the dosage form includes both an immediate release and extended release component. [00512] In one preferred embodiment of the invention, the dosage form includes a capsule within a capsule, each capsule containing a different drug or the same drug intended for treating the same or a different medical condition. In some preferred embodiments, the outer capsule may be an enteric coated capsule or a capsule containing an immediate release formulation to provide rapid plasma concentrations or a rapid onset of effect or a loading dose and the inner capsule contains an extended release formulation. In some preferred embodiments, up to 3 capsules within a capsule are contemplated as part of the invention. In one preferred embodiment of the invention, the dosage form involves one or more tablets within a capsule, wherein the abusable drug is either in the tablet and/or in one of the capsules.
[00513] In one preferred embodiment of the invention, the formulation is ingested orally as a tablet or capsule, preferably as a capsule. In another preferred embodiment of the invention, the formulation is administered bucally. In yet another preferred embodiment of the invention, the formulation is administered sublingually.
[00514] The term "pharmaceutically acceptable salt" as used herein refers to a salt which is toxicologically safe for human and animal administration. Nonlimiting examples of salts include hydrochlorides, hydrobromides, hydroiodides, sulfates, bisulfates, nitrates, citrates, tartrates, bitartrates, phosphates, malates, maleates, napsylates, fumarates, succinates, acetates, terephlhalates, pamoates and pectinates.
[00515] In some embodiments, the abusable drug pharmaceutical composition is a salt or complex of inorganic cation salts, organic salts such primary, secondary, tertiary and quaternary amines include substituted amines In some embodiments, examples of suitable pharmaceutically acceptable salts of abusable drugs include any of the inorganic cation salts such as sodium, potassium, lithium, magnesium, calcium, cesium, ammonia, ferrous, zinc, manganous, aluminum, ferric, and manganic; organic salts with primary, secondary, tertiary and quaternary amines, or mixtures thereof. Examples of such primary, secondary, tertiary and quaternary amines include substituted amines including but not limited to naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and mixtures thereof. More specifically, suitable amines include but are not limited to tromethamine, triethylamine, tripropylamine, dropopizine, 2-dimethylaminoethanol, 2- diethylaminoethanol, lysine, arginine, ornithine, histidine, caffeine, procaine, N-ethylpiperidine, hydrabamine, choline, betaiπe, ethylenediamine, glucosamine, frø-(hydroxymethyl)aminomethane, N-methylglucamine, methylglyc amine, theobromine, piperazine, piperidine, polyamine resins and the like, and mixtures thereof.
[00516] In some embodiments, examples of suitable pharmaceutically acceptable salts of abusable drugs include aminoalcohols chosen from the group consisting of ethanolarnine, 3 -amino- 1 -prop anol, (i?)-l-amino-2- propanol, (S)-I -amino-2-propanol, 2-amino-l,3-propandiol, N-(2- hydroxyethyl)pyrrolidine, D-glucamine and L-prolinόl, D-glucosamine, and iV-methylglucosamine.
[00517] In some embodiments, examples of suitable pharmaceutically acceptable salts of abusable drugs include alkali and alkaline earth metals and salts of an organic nature, such as the salts of basic amino acids.
[00518] It is contemplated that the present invention may be used alone or in combination with other drugs to provide additive, complementary, or synergistic therapeutic effects or for the treatment of entirely different medical conditions.
[00519] Other pharmaceutically active ingredients from various therapeutic classes may also be used in combination with the present invention. They include, but are not limited to decongestants, analgesics, analgesic adjuvants, antidepressants, antipsychotics, anxiolytics, hypnotics, sedatives, anti-ADHD drugs, psychostimulants, drugs to treat urinary incontinence, antihistamines, expectorants, antitussives, diuretics, anti-inflammatory agents, antipyretics, antirheumatics, antioxidants, laxatives, local anesthetics, proton pump inhibitors, motility modifying agents, vasodilators, inotropes, beta blockers, beta adrenergic agonists, drugs to treat asthma and COPD, antiinfectives, antimigraine agents, antihypertensives, antianginal agents, gastric acid reducing agents, anti-ulcer agents, anticoagulants, lipid and cholesterol lowering drugs, anti-diabetic drugs, anti-epileptics, hormones, smooth muscle relaxants, skeletal muscle relaxants, bronchodilators, vitamins, trace minerals, amino acids, biological peptides and drugs to treat various infectious, immunologic disorders, cardiovascular, pulmonary, gastrointestinal, hepatic, biliary, nutritional, metabolic, endocrine, hematologic, oncologic, musculoskeletal, neurologic, psychiatric, genitourinary, gynecologic, obstetric, pediatric, otolaryngogologic, ophthalmic, dermatologic, dental, oral, and genetic disorders, diseases and maladies. The drug being used in combination therapy with the present invention can be administered by any route, including parenterally, orally, topically, transdermally, sublingually, and the like.
[00520] The terms "medical condition", "malady", "disease", "disorder" and
"pathological states" are used interchangeably and are intended to have their broadest interpretation to refer to any physiologic, pathologic or pathophysiologic state in a human that can be prevented, treated, managed or altered to produce a desired, usually beneficial effect.
[00521] In some preferred embodiments, the oral abusable drug is intended to prevent or treat pain. A co-administered drug (in the same or different dosage form, by any route of administration) may be used to provide additive, complementary, superadditive or synergistic therapeutic analgesic effects, including other NSAIDs, NO-NSAIDs, COX-2 selective inhibitors, acetaminophen, nitroparacetamol, nitric oxide donors, tramadol, beta adrenergic agonists, alpha-2 agonists, selective prostanoid receptor antagonists, NO-opioid receptor agonists, local anesthetics, purinergic P2 receptor antagonists, NMDA receptor antagonists, gabapentin, pregabalin, gabapentinoids, ligands of alpha(2)delta subunits of voltage-gated calcium channels, neuronal nicotinic receptor agonists, calcium channel antagonists, sodium channel blockers, superoxide dismutase mimetics, p38 MAP kinase inhibitors, TRPVl agonists, dextromethorphan, dextrorphan, ketamine, glycine receptor antagonists, antidepressants, corticosteroids, and antiepileptics, and any other drugs that can be shown by a person proficient in the art to prevent or treat pain. [00522] Compositions and methods of the present invention provide: (i) abuse deterrence; (ii) extended release; and/or (iii) protection against alcohol dose dumping; and/or (iv) protection against significant changes in bioavailability due to fed or fasted states; and/or (v) simultaneously providing more than one of foregoing (i) to (iv) abuse deterrence and extended release; wherein the dosage form is prepared using compounds selected from the group consisting of: (a) hydrogenated Type I or Type II vegetable oils; (b) polyoxyethylene stearates and distearates; (c) glycerol monostearate; (d) poorly water soluble, high melting point (mp = 45 to 100° C) waxes, and mixtures thereof.
[00523] In some preferred embodiments, the ADER agent of the invention is selected from among hydrogenated Type I or Type II vegetable oils.
[00524] In some preferred embodiments, the ADER agent of the invention is selected from among polyoxyethylene stearates.
[00525] In some preferred embodiments, the ADER agent of the invention is selected from among polyoxyethylene distearates.
[00526] In some preferred embodiments, the ADER agent of the invention is selected from among polyoxyethylene stearates or distearates.
[00527] In some preferred embodiments, the ADER agent of the invention is selected from among poorly water soluble, high melting point (mp = 45 to 100° C) waxes.
[00528] In some preferred embodiments, the ADER agent of the invention is selected from among hydrogenated Type I or Type II vegetable oils; said invention also including an aversive agent.
[00529] In some preferred embodiments, the ADER agent of the invention is selected from among polyoxyethylene stearates; said invention also including an aversive agent.
[00530] In some preferred embodiments, the ADER agent of the invention is selected from among polyoxyethylene distearates; said invention also including an aversive agent.
[00531] In some preferred embodiments, the ADER agent of the invention is selected from among polyoxyethylene stearates or distearates; said invention also including an aversive agent. [00532] In some preferred embodiments, the ADER agent of the invention is selected from among poorly water soluble, high melting point (mp = 45 to
100° C) waxes; said invention also including an aversive agent. [00533] In some preferred embodiments, the ADER agent of the invention excludes hydrogenated Type I vegetable oils. [00534] In some preferred embodiments, the ADER agent of the invention excludes hydrogenated Type II vegetable oils. [00535] In some preferred embodiments, the ADER agent of the invention excludes hydrogenated Type I or Type II vegetable oils. [00536] In some preferred embodiments, the ADER agent of the invention excludes polyoxyethylene stearates. [00537] In some preferred embodiments, the ADER agent of the invention excludes polyoxyethylene distearates. [00538] In some preferred embodiments, the ADER agent of the invention excludes polyoxyethylene stearates or distearates. [00539] In some preferred embodiments, the ADER agent of the invention excludes poorly water soluble, high melting point (mp = 45 to 100° C) waxes. [00540] In some preferred embodiments, the ADER agent of the invention excludes poorly water soluble, high melting point (mp = 50 to 100° C) waxes. [00541] In some preferred embodiments, the ADER agent of the invention excludes poorly water soluble, high melting point (mp = 60 to 100° C) waxes. [00542] In some preferred embodiments, the ADER agent of the invention excludes poorly water soluble, high melting point (mp = 70 to 85° C) waxes. [00543] In some preferred embodiments, the ADER agent of the invention excludes poorly water soluble, high melting point (mp = 75 to 90° C) waxes. [00544] In some preferred embodiments, the ADER agent of the invention excludes poorly water soluble, high melting point (mp = 70 to 100° C) waxes. [00545] In a particularly preferred embodiment of the invention, the dosage form includes two or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate;
(iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes. [00546] In a most preferred embodiment of the invention, the dosage form includes two or more compounds selected from at least two categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes.
[00547] In some preferred embodiments, the dosage form includes hydrogenated Type I or Type II vegetable oils in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
[00548] In some preferred embodiments, the dosage form includes polyoxyethylene stearates in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg. [00549] In some preferred embodiments, the dosage form includes polyoxyethylene distearates in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
[00550] hi some preferred embodiments, the dosage form includes glycerol monostearate in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
[00551] In some preferred embodiments, the dosage form includes poorly water soluble, high melting point (mp = 45 to 100° C) waxes in an amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
[00552] In some preferred embodiments, the dosage form includes two or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes in total (i.e. cumulative) amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
[00553] In some preferred embodiments, the dosage form includes the dosage form includes two or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water, soluble, high melting point (mp = 45 to 100° C) waxes in total (i.e. cumulative) amount that is less than about 1200 mg, or less than about 100 mg, or less than about 900 mg, or less than about 800 mg, or less than about 700 mg, or less than about 600 mg, or less than about 550 mg, or less than about 500 mg, or less than about 400 mg, or less than about 375 mg, or less than about 350 mg, or less than about 325 mg, or less than about 300 mg, or less than about 275 mg, or less than about 250 mg, or less than about 225 mg, or less than about 200 mg, or less than about 175 mg, or less than about 150 mg, or less than about 125 mg, or less than about 100 mg, or less than about 90 mg, or less than about 80 mg, or less than about 70 mg, or less than about 60 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 20 mg, or less than about 10 mg.
[00554] Representative examples of hydrogenated vegetable oils of the present invention include, without limitation, hydrogenated cottonseed oil (e.g., Akofine®; Lubritab®; Sterotex® NF), hydrogenated palm oil (Dynasan® P60; Softisan® 154), hydrogenated soybean oil (Hydrocote®; Lipovol HS-K®; Sterotex® HM) and hydrogenated palm kernel oil (e.g., Hydrokote® 112).
[00555] Representative examples of polyoxyethylene stearates and distearates of the present invention include, without limitation, Polyoxyl 2, 4, 6, 8, 12, 20, 30, 40, 50, 100 and 150 stearates (e.g., Hodag® DGS; PEG-2 stearate; Acconon® 200-MS; Hodag® 20-S; PEG-4 stearate; Cerasynt® 616; Kessco® PEG 300 Monostearate; Acconon® 400-MS; Cerasynt® 660; Cithrol® 4MS; Hodag® 60-S; Kessco® PEG 600 Monostearate; Cerasynt® 840; Hodag 100-S; Myrj® 51; PEG-30 stearate; polyoxyethylene (30) stearate;Crodet® S40; E431; Emerest® 2672; Atlas G-2153; Crodet® S50) and polyoxyl 4, 8, 12, 32 and 150 distearates (e.g, Lipo-PEG® 100-S; Myrj® 59; Hodag® 600-S; Ritox® 59; Hodag® 22-S; PEG-4 distearate; Hodag® 42-S; Kessco® PEG 400 DS; Hodag® 62-S; Kessco® PEG 600 Distearate; Hodag® 154-S; Kessco® PEG 1540 Distearate; Lipo-PEG® 6000-DS; Protamate® 6000-DS).
[00556] Representative examples of poorly water soluble, high melting point
(mp = 45 to 100° C) waxes of the present invention include, without limitation: (i) animal waxes; (ii) insect waxes; (iii) vegetable waxes; (iv) mineral waxes; (v) petroleum waxes; (vi) synthetic waxes; (vi) nonionic emulsifying waxes or cetomacrogol emulsifying wax (e.g., Collone TV/™; Crodex N™; Emulgade 1000NI™; Permulgin D™; Polawax™; Ritachol 2000; T- Wax™); (vii) anionic emulsifying wax (e.g., Collone HV™; Crodex A™; Cyclonette wax; Lanette wax SX™BP); (Vzϊz^carnauba wax (also known as Brazil wax; caranda wax; E903); (ix) microcrystalline wax (also known as amorphous wax; E907; petroleum ceresin; petroleum wax (microcrystalline)); (x) yellow wax (e.g., yellow beeswax; Apifil™; E901; refined wax]; (xi) white wax (bleached wax; E901); (xii) cetyl esters wax (e.g., cera cetyla; Crodamol SS™; Cutina CP™; Liponate SPS™; Protachem MST™; Ritaceti™; Ritachol SS™; spermaceti wax replacement; Starfol wax CG™; Synaceti 116™; synthetic spermaceti); (xiii) hydrogenated castor oil (e.g., Castorwax™; Castorwax MP 70™; Castorwax MP 80™; Croduret™; Cutina HR™; Fancol™; Simulsol 1293™); (xiv) /αnolin alcohols (e.g., Cholesterol; lanolin; lanolin, hydrous; petrolatum and lanolin alcohols; mineral oils); (xv) lanolin (e.g., cera lanae; E913; lanolina; lanolin anhydrous; Protalan anhydrous; purified lanolin; refined wool fat); (xvi) glyceryl palmitostearate; (xvii) cetostearyl alcohol (e.g., cetearyl alcohol; Crodacol CS90™; Lanette O™; Tego Alkanol 1618™; Tego Alkanol 6855™); (xviii) beeswax. ] In some embodiments, the dosage form is devoid of animal waxes. In other embodiments, the dosage form is devoid of insect waxes. In other embodiments, the dosage form is devoid of vegetable waxes. In other • embodiments, the dosage form is devoid of mineral waxes. In other embodiments, the dosage form is devoid of petroleum waxes. In other embodiments, the dosage form is devoid of synthetic waxes. In other embodiments, the dosage form is devoid of nonionic emulsifying waxes or cetomacrogol emulsifying wax. hi other embodiments, the dosage form is devoid of anionic emulsifying wax. In other embodiments, the dosage form is devoid of carnauba wax. In other embodiments, the dosage form is devoid of microcrystalline wax. In other embodiments, the dosage form is devoid of yellow wax. In other embodiments, the dosage form is devoid of white wax. hi other embodiments, the dosage form is devoid of cetyl esters wax. In other embodiments, the dosage form is devoid of hydrogenated castor oil. In other embodiments, the dosage form is devoid of lanolin alcohols, hi other embodiments, the dosage form is devoid of lanolin, hi other embodiments, the dosage form is devoid of glyceryl palmitostearate. hi other embodiments, the dosage form is devoid of cetostearyl alcohol, hi other embodiments, the dosage form is devoid of beeswax. [00558] In one preferred embodiment of the present invention, the abusable drug is combined with beeswax, hydroxypropyl methyl cellulose (e.g, HPMC Kl 5M), silicon dioxide (alone or in combination with Al2O3; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84).
[00559] In one preferred embodiment of the present invention, the abusable drug is combined with hydrogenated cottonseed oil (e.g., Sterotex® NF), hydroxypropyl methyl cellulose (e.g, HPMC Kl 5M), fractionated coconut oil and silicon dioxide (alone or in combination with AI2O3; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84).
[00560] In another preferred embodiment of the present invention, the abusable drug is combined with glycerol monostearate (e.g., Cithrol® GMS), hydroxypropyl methyl cellulose (e.g, HPMC KlOOM) and silicon dioxide (alone or in combination with Al2O3; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84).
[00561] In yet another preferred embodiment of the present invention, the abusable drug is combined with hydrogenated palm kernel oil (e.g., Hydrokote® 112), hydroxypropyl methyl cellulose (e.g, HPMC Kl 5M) and silicon dioxide (alone or in combination with Al2O3; e.g, Aerosil®, Aerosil® 200, Aerosil® COK84).
[00562] In one preferred embodiment of the present invention, release rate modifiers may be incorporated. Release rate modifiers can also have additional useful properties that optimize the formulation.
[00563] In one preferred embodiment of the present invention, also included are cellulose and cellulose derivatives including, without limitation cellulose acetate, microcrystalline cellulose, powdered cellulose, cellulose acetate phthalate, hydroxyethyl cellulose, silicified microcrystalline cellulose, hydroxypropyl cellulose, hydroxyethylmethyl cellulose, low-substituted hydroxypropyl cellulose, carboxymethylcellulose, carboxymethylcellulose calcium, hypromellose acetate succinate, hypromellose phthalate and ethylcellulose.
[00564] In one preferred embodiment of the present invention, also included are coconut oil products, including without limitation, coconut oil, fractionated coconut oil, cetyl alcohol, lauric acid and medium chain triglycerides (e.g., Bergabest; caprylic/capric triglyceride; Captex 300; Captex 355; Crodamol GTC/C; glyceryl tricaprylate/caprate; Labrafac CC; MCT oil; Miglyol 810™; Miglyol 812™; Myritol; Neobee M5™; Nesatol™; oleum neutrale; oleum vegetable tenue; thin vegetable oil; Waglinol 3/9280™). In a most preferred embodiment, the coconut oil is fractionated coconut oil.
[00565] In one preferred embodiment of the present invention, hydroxypropyl methyl cellulose (e.g, HPMC Kl 5M) may be incorporated.
[00566] A variety of agents may be incorporated into the ADER invention as thixotropes (e.g., fumed silicon dioxides, Aerosil®, Aerosil® COK84, Aerosil® 200, etc.). Thixotropes enhance the pharmaceutical formulations of the invention by increasing the viscosity of solutions during attempted extraction, complementing the action of HPMCs. They may also provide a tamper resistance by helping to retain the structure of dosage units that have been heated to temperatures greater than the melting point of the base excipient (Aerosils are unaffected by heat).
[00567] As described above, the present invention can include one or more
ADER agents. Any amount of ADER may be used. In some embodiments, the total amount of ADER agent is about 5 to about 98 percent, preferably 7 to 90 percent and more preferably 10 to 85 percent on a dry weight basis of the composition.
[00568] In one preferred embodiment, the ADER can prevent less than or equal to about 98%, 90%, 80% 75%, 60%, 50%, 45%, 40%, 33%, 30%, 25%, 15%, 10%, 8%, 5%, or 2% of the total amount of drug in a dosage form from being recovered from a solvent in contact with a dosage form of the present invention.
[00569] In some preferred embodiments, the dosage form is devoid of hydrogenated Type I vegetable oils. In other embodiments, the dosage form is devoid of hydrogenated Type II vegetable oils, m other embodiments, the dosage form is devoid of polyoxyethylene stearates. In other embodiments, the dosage form is substantially of polyoxyethylene distearates; in other embodiments, the dosage form is substantially of glycerol monostearate. In other embodiments, the dosage form is substantially of poorly water soluble, high melting point (mp = 45 to 100° C) waxes.
[00570] In preferred embodiments, the abuse deterrent, extended release, alcohol dose dumping protective and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a hydrogenated Type I vegetable oil. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a hydrogenated Type II vegetable oil. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a hydrogenated Type I or Type II vegetable oil. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a polyoxyethylene stearate. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a polyoxyethylene distearate. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, each a glycerol monostearate. In other embodiments, the abuse deterrent, extended release and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents,, each a wax.
[00571] In some particularly preferred embodiments, the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) a hydrogenated Type I vegetable oil; (ii) a hydrogenated Type II vegetable oil; (iii) a polyoxyethylene stearate; (iv) a polyoxyethylene distearate; (v) a glycerol monostearate; and (vi) a wax.
[00572] In some particularly preferred embodiments, the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) a hydrogenated Type I vegetable oil; (ii) a hydrogenated Type II vegetable oil; (iii) a polyoxyethylene stearate; (iv) a polyoxyethylene distearate; (v) a glycerol monostearate; and (vi) a wax, said dosage form having a melting point > 50°C, or > 55°C, or > 600C, or > 65°C, or > 700C, or > 75°C, or > 800C, or > 85°C, or > 9O0C, or between 50 and 600C, or between 55 and 65°C, or between 60 and 7O0C, or between 65 and 75°C, or between 70 and 800C, or between 75 and 850C, or between 80 and 900C, or between 90 and 1000C, or between 50 and 1000C, or between 60 and 1000C, or between 70 and 1000C, or between 80 and 1000C, or between 60 and 800C, or between 60 and 900C, or between 65 and 85°C, or between 70 and 900C.
[00573] In other particularly preferred embodiments, the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) hydrogenated Type I or Type II vegetable oil; (ii) polyoxyethylene stearate or distearate; (iii) a glycerol monostearate; and (iv) a wax.
[00574] In other particularly preferred embodiments, the abuse deterrent, extended release, resistance against alcohol dose dumping when formulated as extended release, and/or abuse deterrent plus extended release dosage form of the invention comprises at least two ADER agents, selected from at least two groups, comprising: (i) hydrogenated Type I or Type II vegetable oil; (ii) polyoxyethylene stearate or distearate; (iii) a glycerol monostearate; and (iv) a wax, said dosage form having a melting point > 500C, or > 55°C, or > 600C, or > 65°C, or > 700C, or > 75°C, or > 800C, or > 85°C, or > 900C, or between 50 and 600C, or between 55 and 65°C, or between 60 and 700C, or between 65 and 75°C, or between 70 and 800C, or between 75 and 85°CS or between 80 and 9O0C, or between 90 and 1000C, or between 50 and 1000C, or between 60 and 1000C, or between 70 and 1000C, or between 80 and 1000C, or between 60 and 800C, or between 60 and 900C, or between 65 and 85°C, or between 70 and 900C.
[00575] The present invention can also optionally include other ingredients to enhance dosage form manufacture from a pharmaceutical composition of the present invention and/ or alter the release profile of a dosage form including a pharmaceutical composition of the present invention.
[00576] Some embodiments of the present invention include one or more pharmaceutically acceptable fillers, diluents, glidants and lubricants of various particle sizes and molecular weights.
[00577] The dosage form according to the invention may also comprise a coating which is resistant to gastric juices and dissolves as a function of the pH value of the release environment. By means of this coating, it is possible to ensure that, when correctly administered, the dosage form according to the invention passes through the stomach undissolved and the active ingredient is only released in the intestines.
[00578] In some preferred embodiments, the dosage form may include a surfactant ingredient to impart suitable formulation characteristics to the composition. Surfactants may be hydrophilic preferably selected from the group consisting of non-ionic hydrophilic surfactants and anionic hydrophilic surfactants or the surfactant may have hydrophobic properties. Examples of non-ionic hydrophilic surfactants are polyoxyethylene sorbitan esters, cremophores and poloxamers. Examples of anionic surfactants are sodium lauryl sarcosinate, docusate and pharmaceutically acceptable docusate salts. Also a mixture of these surfactants can be used.
[00579] The formulation optionally comprises auxiliary materials. Examples of these auxiliary materials (or pharmaceutically acceptable excipients) are (i) Binders such as acacia, alginic acid and salts thereof, cellulose derivatives, methylcellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, magnesium aluminum silicate, polyethylene glycol, gums, polysaccharide acids, bentonites, hydroxypropyl methylcellulose, gelatin, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer, crospovidone, povidone, polymethacrylates, hydroxypropylmethylcellulose, hydroxypropylcellulose, starch, pregelatinized starch, ethylcellulose, tragacanth, dextrin, microcrystalline cellulose, sucrose, or glucose, and the like; (ii) Disintegrants such as starches, pregelatinized corn starch, pregelatinized starch, celluloses, cross-linked carboxymethylcellulose, crospovidone, cross-linked polyvinylpyrrolidone, a calcium or a sodium alginate complex, clays, alginates, gums, or sodium starch glycolate, and any disintegration agents used in tablet preparations; (iii) Filling agents such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like; (iv) Stabilizers such as any antioxidation agents, reducing agents, buffers, or acids, sodium citrate, ascorbyl palmitate, propyl gallate, ascorbic acid, vitamin E, sodium bisulfite, butylhydroxyl toluene, BHA, acetylcysteine, monothioglycerol, phenyl- alpha-nathylamine, lecithin, EDTA, and the like.; (v) Lubricants such as magnesium stearate, calcium hydroxide, talc, colloidal silicon dioxide, sodium stearyl fumarate, hydrogenated vegetable oil, stearic acid, glyceryl behenate, magnesium, calcium and sodium stearates, stearic acid, talc, waxes, Stearowet, boric acid, sodium benzoate, sodium acetate, sodium chloride, DL-leucine, polyethylene glycols, sodium oleate, or sodium lauryl sulfate, and the like; (vi) Wetting agents such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium oleate, or sodium lauryl sulfate, and the like; (vii) Diluents such lactose, starch, mannitol, sorbitol, dextrose, microcrystalline cellulose, dibasic calcium phosphate, sucrose-based diluents, confectioner's sugar, monobasic calcium sulfate monohydrate, calcium sulfate dihydrate, calcium lactate trihydrate, dextrates, inositol, hydrolyzed cereal solids, amylose, powdered cellulose, calcium carbonate, glycine, or bentonite, and the like; (viii) Anti-adherents or glidants such as talc, corn starch, DL-leucine, sodium lauryl sulfate, and magnesium, calcium, or sodium stearates, and the like; (ix) Pharmaceutically compatible carriers such as acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, malto dextrin, glycerin, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, or pregelatinized starch, and the like; (x) Other pharmaceutical excipients including polymers, hydrogels, silicon dioxide, ion exchange resins, cellulose acetate butyrate, carbohydrate polymers, organic acids of carbohydrate polymers caprylic/capric triglyceride, isopropyl myristate, ethyl oleate, triethyl citrate, dimethyl phthalate, and benzyl ben- zo ate. The pharmaceutical compositions and dosage form of the invention may further contain one or more pharmaceutically acceptable excipients. When used in the present invention, pharmaceutically acceptable excipients can play a small or significant role in the behavior of the dosage form, depending on the choice of excipient, quantity of excipient and interaction with other constituents of the dosage form and the gastrointestinal tract. Pharmaceutically acceptable excipients are well known in the art and include, without limitation, excipients referenced in the FDA EAFUS database (http://vmxfsan.fda.gov/~dms/eafus.html); FDA Food Additives Status List (http://www.cfsan.fda.gov/~dms/opa-appa.html); FDAGRAS list and database; FDA Color Additive Status List
(http://www.cfsan.fda.gov/~dms/opa-appc.html); FDA Inactive Ingredients Database (http://www.accessdata.fda.gov/scripts/cder/iig/index.cfm); Rowe, Sheskey and Owen, Handbook of Pharmaceutical Excipients, APhA Publications; 5th edition (2006); Remington: The Science and Practice of Pharmacy, 21st ed, Lippincott Williams & Wilkins (2005); United States Pharmacopeia-National Formulary (USP-NF), (USP 30 - NF 25, 2007), the International Programme on Chemical Safety (http://www.inchem.org/) and Health Canada's List of Acceptable Non-medicinal Ingredients (http://www.hc-sc.gc.ca/dhp-mps/prodnatur/legislation/docs/nmi- imn_listl_e.html), all hereby incorporated by reference in their entirety. [00581] The dosage form according to the invention may also comprise a coating which is resistant to gastric juices and dissolves as a function of the pH value of the release environment. [00582] By means of this coating, it is possible to ensure that, when correctly administered, the dosage form according to the invention passes through the stomach undissolved and the active ingredient is only released in the intestines. [00583] In one preferred embodiment of the invention, the formulation is ingested orally as a tablet or capsule, preferably as a capsule. In another preferred embodiment of the invention, the formulation is administered bucally. In yet another preferred embodiment of the invention, the formulation is administered sublingually. [00584] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to prevent or minimizing excessive peak concentrations (dose dumping) of therapeutic doses of extended release abusable drugs for medical purposes, when they are co-ingested with alcohol. [00585] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve an extended release formulation of abusable drugs. [00586] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve an abuse deterrent formulation of abusable drugs. [00587] hi one preferred embodiment, the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release and abuse deterrence for abusable drugs. [00588] hi one preferred embodiment, the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release and abuse deterrence for abusable drugs, using substantially the same excipients. [00589] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve abuse deterrence, without the use of aversive agents. [00590] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to simultaneously achieve extended release and abuse deterrence, without the use of aversive agents.
[00591] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release formulation and an abuse deterrence formulation, using substantially the same ADER agents.
[00592] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents.
[00593] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention providing abuse deterrence without the use of aversive agents.
[00594] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention using substantially the same ADER agents to achieve the foregoing (i) to (v).
[00595] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention using substantially the same ADER agents to achieve the foregoing (i) to (v); said invention providing abuse deterrence without the use of aversive agents.
[00596] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and at least two ADER agents.
[00597] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and at least two ADER agents; said invention providing abuse deterrence without the use of aversive agents.
[00598] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to achieve the following properties: (i) abuse deterrence; and/or (ii) extended release; and/or (iii) resistance against alcohol dose dumping; and/or (iv) resistance against alcohol dose dumping when formulated as extended release; and/or (v) more than one of the foregoing [(i) to (iv)] properties; said invention comprising one or more abusable drugs and at least two ADER agents; said invention using substantially the same ADER agents to achieve the foregoing (i) to (v).
[00599] In one preferred embodiment, the invention provides for methods and pharmaceutical compositions to simultaneously achieve an extended release formulation and an abuse deterrence formulation, using substantially the same
ADER agents without the use of aversive agents. [00600] It is understood that each of the various embodiments of methods and pharmaceutical compositions described herein may be used alone or in conjunction with one or more or all of the various embodiments described herein. [00601] Additionally, it is understood that each of the various embodiments of the pharmaceutical compositions described herein may be used with each of the various embodiments of the described method of the present invention as described herein.
Determination of Biologic Effects in Humans
[00602] The pharmacologic effects of the pharmaceutical compositions of the present invention can be evaluated using methods well established in the art. The choice of method will depend, among other things, on: (i) the abusable drug and (ii) the therapeutic use to which the abusable drug is applied (i.e., the disease, disorder, or symptom(s) being treated with the abusable drug). Some abusable drugs, for example, cannabinoid agonists, stimulant drugs, or benzodiazepines have multiple therapeutic applications. Additionally, certain evaluations may be conducted in healthy subjects, recreational drug users or drug addicts. A wide variety of clinical states and study designs may be used to evaluate the therapeutic effects of intact and tampered dosage forms of the invention. This invention therefore contemplates the use of test methods other than those specifically disclosed herein, including those which may hereafter become known to the art to be capable of performing the necessary functions. Sample sizes in the studies are sufficient to demonstrate the objectives of the testing. A non-limiting list of methods to evaluate the analgesic and other effects of the invention is provided below:
Third Molar Extraction Model
[00603] Male and female patients with acute postsurgical pain following the removal of one or more bony impacted third molars are participants. Within 4 to 6 hours after completion of surgery, patients who are experiencing moderate or severe pain, as measured by a visual analog pain intensity scale (VAS > 50 mm) and by a categorical pain intensity scale (moderate or severe pain descriptor), and who meet all other inclusion/exclusion criteria are admitted to the study. Patients are randomly assigned to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Both single and multiple (repeated) dose studies may be conducted. Pain intensity (VAS and categorical), pain relief (categorical) and whether pain is half-gone is recorded by the patient under the supervision of the investigator study coordinator at the various time points: Baseline (0 hour - pain intensity only), 15, 30 and 45 minutes, and at 1, 1.5, 2, 3, 4, 5, 6, 7, 8 and 12 hours after administration of study medication, and immediately prior to the first rescue dose. Sedation and nausea may be evaluated using VAS or categorical scales. Time to onset of perceptible and meaningful pain relief is evaluated using the two stopwatch method. Patients record their global evaluation of study medication at the completion of the 8-hour assessment or at the time of first rescue medication use. Efficacy endpoints include Total Pain Relief (TOTPAR), Sum of Pain Intensity Difference (SPID) and Sum of Pain Relief Intensity Difference (SPRID) at various time points, Time to First Rescue, Time Specific Pain Intensity Difference (PID), Time Specific Pain Relief (PR), Peak Pain Intensity Difference (PPID)5 Peak Pain Relief (PPR), Time to Confirmed Perceptible Pain Relief (stopwatch), Time to Meaningful Pain Relief (stopwatch), Patient Global Evaluation, Time to Change in Categorical PID > 1, Percent Change in Pain Intensity Score from Baseline, Mean Change in Pain Intensity Score From Baseline, Percent Change in Pain Relief Score from Baseline, Mean Change in Pain Relief Score From Baseline, Percent of Responders, Number of Patients Needed to Treat to Obtain One Patient with > 50% Response (NNT).
Bunionectomy Surgery [00604] Male or female patients requiring primary unilateral first metatarsal bunionectomy surgery alone or with ipsilateral hammertoe repair (without additional collateral procedures) under regional anesthesia (Mayo block) are participants.
[00605] Patients who experience moderate or severe pain on a categorical scale
(moderate or severe descriptor) and on a visual analog pain intensity scale (VAS; >50 mm) within 6 hours following completion of bunionectomy surgery are randomly assigned to receive the dosage form of the invention given intact or placebo In some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Both single and multiple (repeated) dose studies may be conducted. Patients are encouraged to wait at least 60 minutes before requesting remedication for pain. At the completion of the single-dose phase (S hours) or at first request for remedication (whichever is earlier), patients enter into a multiple-dose phase lasting approximately 72 hours. During the multiple dose phase patients receive study medication or placebo at a fixed dose interval (e.g., every 8, 12 or 24 hours). Once the multiple dose phase of the study has begun, patients experiencing pain between scheduled doses of study medication are provided access to supplemental open-label (rescue) analgesia. Patients whose pain cannot be adequately managed on a combination of study medication and rescue medication or who develop unacceptable side effects during the study are discontinued from " further study participation and their pain managed conventionally.
[00606] Pain intensity (VAS and categorical), pain relief (categorical) and whether pain is half-gone is recorded by the patient under the supervision of the investigator study coordinator at representative time points, e.g., Baseline (pain intensity only), 15, 30 and 45 minutes and 1, 1.5, 2, 3, 4, 5, 6, 7 and 8 hours after administration of study medication and immediately prior to the first remedication. Sedation and nausea may be evaluated using VAS or categorical scales. Time to onset of perceptible and meaningful pain relief is evaluated using the double-stopwatch method. Patients complete a global evaluation of study medication at the completion of the 8-hour assessment or just prior to the first remedication. Following completion of the single-dose phase (8 hours or just prior to first remedication, if < 8 hours), patients begin the multiple dose phase of the study. During the multiple dose phase, patients record their overall pain intensity since the previous scheduled dose, their current pain intensity and a patient global, immediately prior to each scheduled dose of study medication and at early termination.
[00607] Measures of efficacy in the single-dose phase include Sum of Pain
Intensity Difference (SPID), Total Pain Relief (TOTPAR), Sum of Pain Relief Intensity Difference (SPRID), Time to First Remedication, Time Specific Pain Intensity Difference (PID), Time Specific Pain Relief (PR), Peak Pain Intensity Difference (PPJD), Peak Pain Relief (PPR), Time to Confirmed Perceptible Pain Relief (stopwatch), Time to Meaningful Pain Relief (stopwatch), Patient Global Evaluation, Time to Change in Categorical PID > 1, Percent Change in Pain Intensity Score from Baseline, Mean Change in Pain Intensity Score From Baseline, Percent Change in Pain Relief Score from Baseline, Mean Change in Pain Relief Score From Baseline, Percent of Responders, Number of Patients Needed to Treat to Obtain One Patient with > 50% Response (NNT).
[00608] Measures of efficacy in the multiple-dose phase include the time specific overall pain intensity, current pain intensity and patient global at the time of scheduled remedication, the average of overall pain intensity, current pain intensity and patient global over 0-24, 24-48 and 48-72 and number of doses of rescue analgesic over 0-24, 24-48 and 48-72 and 0-72 hours.
Chronic Pain of Osteoarthritis
[00609] The analgesic efficacy of the invention may be demonstrated in single or repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. In repeated dose studies, typically, patients who meet the American College of Rheumatology criteria for knee and/or hip OA are washed off their analgesics for 2 to 7 days to allow for pain of moderate to severe intensity to return. Once a stable baseline pain score is established, patients are randomized to treatment, usually for a period of 1 to 12 weeks. Pain, joint stiffness and physical function can be measured with a multidimensional instrument, such as the WOMAC, quality of life with the SF- 12 or SF-36 and adverse events with a non-directed questionnaire at baseline and at post-baseline return visits. Response to pain, stiffness, physical function, quality of life and adverse events are calculated as change from baseline and compared between treatments. Sedation and nausea may be evaluated using VAS or categorical scales.
Migraine
[00610] The analgesic efficacy of the invention may be demonstrated in single or repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Patients with migraine headaches are typically evaluated in prospective, randomized, double-blind, parallel group, single-dose studies. Crossover studies are also possible. The study population consists of male and non-pregnant female subjects, 18 to 65 years of age with a primary headache diagnosis of either migraine attack without aura or migraine attack with aura, as diagnosed according to the International Classification of Headache Disorders-2 criteria. To qualify, the subject must typically have a history, on average, of at least one migraine attack per month, but an average of no more than 6 migraine attacks each month during the past year. Using a headache diary subjects are instructed to treat and evaluate the headache pain and symptoms associated with one eligible migraine attack, with or without aura, with at least moderate headache pain intensity. Eligible subjects are randomly assigned to receive the drug to treat one migraine attack, with or without aura, with headache pain of at least moderate pain intensity as determined by them migraine questionnaire they are asked to take a single dose of study drug, according to their randomized treatment assignment. Headache pain intensity, nausea, photophobia, phonophobia, vomiting, and ability to function are assessed at baseline, 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, 8, 16 and 24 hours post-dose. In addition, the recurrence of pain and use of any rescue mediation is documented. Primary efficacy variables typically consist of the percent of subjects who are without: (i) pain; (ii) nausea; (iii) photophobia and, (iv) phonophobia, each at 2 hours post-dosing. Secondary efficacy variables typically consist of headache pain intensity and associated symptoms at each evaluation time point, incidence of vomiting, patient function, sum of pain intensity difference at each evaluation time (SPED), percent of subjects who experience headache recurrence up to 24 hours, and the median time to recurrence. Sedation may be evaluated using VAS or categorical scales. Recurrence is defined as the reduction in pain from moderate or severe pain to none at 2 hours after taking study drug, followed by: (i) an increase to mild, moderate or severe pain within 24 hours after taking the study drug, or (ii) consuming a rescue medication within 24 hours after taking the study drug.
Postherpetic Neuralgia
[00611] The analgesic efficacy of the invention may be demonstrated in repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Patients with a history of postherpetic neuralgia > 3 months and pain of at least moderate intensity are enrolled in the study. Patients with hypersensitivity to study medications, a history of drug or alcohol abuse and significant pain of alternate etiology are generally excluded. Patients meeting study eligibility criteria are "washed off their analgesics in some embodiments, generally for 2 to 7 days to allow for pain of moderate to severe intensity to return. Once a stable baseline pain score is established, patients are randomized to treatment, usually for a period of 4 to 12 weeks. Pain intensity is assessed one to several times a day and in some cases only once weekly using VAS, categorical or numerical rating scales. Various dimensions of neuropathic pain may be assessed, including steady pain (ongoing pain), brief pain (paroxysmal pain) and skin pain (allodynia). Pain may also be assessed at scheduled clinic study visits. Pain may also be assessed using standardized pain scales such as the Neuropathic Pain Scale (Galer et al., Neurology 1997;48:332-8), the Neuropathic Pain Symptom Inventory (Bouhassira et al., Pain 2004;108:248-57), interference measures of the Brief Pain Inventory (Cleeland, CRC Press, 1991:293-305 and Ann Acad Med Singapore 1994;23: 129-38) or the McGiIl Pain Questionnaire Short- Form (Melzack, Pain 1987;30:191-7). Patient global assessment may be measured using a number of available tools, for example Patient Global Impression of Change (Farrar et al., Pain 2001 ;94: 149-580). Quality of life may similarly be assessed using number of available tools, for example the SF-36, SF-12 or SF-8. Examples of randomized, placebo or active studies conducted in postherpetic neuralgia are known in the art (e.g., Watson and Babul, Neurology 1998;50:1837-41; Sabatowski et al., Pain. 2004;109:26-35; Rowbotham et al., JAMA. 1998;280: 1837-42). Adverse events may be assessed using a non-directed questionnaire, a symptom checklist or specific queries on adverse signs and symptoms. Response to pain, function, quality of life and adverse events are calculated as change from baseline and compared between treatments. Sedation and nausea may be evaluated using VAS or categorical scales.
Attention-deficit/hyperactivity disorder (ADHD')
[00612] The efficacy and safety of the invention may be demonstrated in repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Patients are usually titrated to optimal doses. Adults or children with a history of ADHD meeting DSM-IV-TR criteria for ADHD are enrolled in the study. Patients with hypersensitivity to study medications, a history of drug or alcohol abuse and significant co-morbid pathology that could confound ADHD assessment are generally excluded. Outcome measures for adult ADHD studies may include the Wender-Reimherr Adult Attention Deficit Disorder Scale (WRAADDS), the adult ADHD-Rating Scale (ADHD- RS), the Conners' Adult ADHD Rating Scale Short Version Self-Report (CAARS-S-S), and the Clinical Global Impressions-Improvement scale (CGI- I). Outcome measures in children may include the teacher-completed Swanson, Nolan, and Pelham Rating Scale (Teacher SNAP), parent-completed SNAP (Parent SNAP)5 Clinical Global Impressions Scale-Improvement (CGI- I) score, Math Test performance, the Teacher and Parent ADHD Rating Scale, the Conners' ADHD/DSM-IV Scales for teachers (CADS-T) and for parents (CADS-P). Patients are randomized to treatment, usually for a period of 2 to 12 weeks. Adverse events may be assessed using a non-directed questionnaire, a symptom checklist or specific queries on adverse signs and symptoms. Response may be calculated as change from baseline and compared between treatments.
Insomnia
[00613] The efficacy and safety of the invention may be demonstrated in repeated dose randomized double-blind, controlled studies. Patients are randomized to receive the dosage form of the invention given intact or placebo, in some preferred embodiments, and the dosage form of the invention given intact or dosage form of the invention given in tampered form in other embodiments. Aged 21 to 69 years meeting DSM-IV-TR criteria for primary insomnia and reporting less than 6.5 hours of sleep per night, and/or a sleep latency of more than 30 minutes each night for at least 1 month before screening are enrolled in the study. Patients with hypersensitivity to study medications, a history of drug or alcohol abuse and significant co-morbid pathology that could confound sleep assessment are generally excluded. Outcome measures for may include Efficacy and next-morning effects may be evaluated via polysomnography, Digit Symbol Substitution Test and self- report. Study endpoints may include sleep latency, total sleep time, number of awakenings, wake time after sleep onset, quality of sleep and next-day ratings of ability to function, daytime alertness, and sense of physical well-being. Patients are randomized to treatment, usually for a period of 2 to 6 weeks. Adverse events may be assessed using a non-directed questionnaire, a symptom checklist or specific queries on adverse signs and symptoms. Response may be calculated as change from baseline and compared between treatments.
[00614] The preparation of oral immediate release dosage forms is well known in the art - see Remington: the science of Pharmacy Practice, 21st Edition, 2006, Lippincott, Williams & Wilkins, Baltimore, MD; Pharmaceutical Preformulation and Formulation: A Practical Guide from Candidate Drug Selection to Commercial Dosage Form. Gibson, M (ed). CRC Press, 2001; Niazi, S. Handbook of Pharmaceutical Manufacturing Formulations: Uncompressed Solid Products (Volume 2 of 6), CRC Press, 2004; Niazi, S. Handbook of Pharmaceutical Manufacturing Formulations: Compressed Solid Products (Volume 1 of 6), CRC Press, 2004; Mollet, H, Grubenmann A, Payne H. Formulation Technology: Emulsions, Suspensions, Solid Forms, Wiley-VCH, 2001; Niazi S and Niazi SK, Pharmaceutical Capsules, 2nd Ed., Podczeck, F and Jones BE (eds)., Pharmaceutical Press, 2004, London (all of which are hereby incorporated by reference). A majority of oral dosage forms commercially available world wide are formulated as immediate release products.
Evaluating Effects Fed and Fasted State on Oral Bioavailability
[00615] The effects of fed and fasted state of the invention, and in particular, their effects on Cm8x, AUCo-t and AUCo-00, can be evaluated using methods well know in the art. More specifically, the invention relies on the methods contained in the document entitled: "Guidance for Industry: Food-Effect Bioavailability and Fed Bioequivalence Studies: Study Design, Data Analysis and Labeling", Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research, October, 2001, which is here incorporated by reference. Additional guidance on the conduct of bioavailability studies is found in "Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products-General Considerations", Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research, July 2002, which is here incorporated by reference.
Tamper Resistance and Tamper Deterrence Testing
[00616] The popularity of immediate release formulations of abusable drugs among drug addicts and recreational drug users is in part due to the mood altering and reinforcing effects of the drug. The popularity of extended release formulations of abusable drugs among drug addicts and recreational drug users is in part due to the pharmacologic properties of the drug therein (e.g., mood altering and reinforcing effects) and in part due to the large amount of drug per tablet or capsule (e.g., a 12 or 24 hour supply). For example commercially available immediate release abusable drugs tablets and capsules are usually administered every 4 to 6 hours and they release their dose into the systemic circulation over one to two hours. New, extended release formulations are designed to gradually release their much larger abusable drugs content over a 12 or 24-hour period. Most recreational drug users and addicts have a unit of use which is one tablet or capsule. The 12 or 24-hour supply of an abusable drugs typically contained in one extended release tablet or capsule, instead of in 4 to 6 tablets or capsules means that there is a greater risk that such formulations may be highly sought by drug addicts and recreational drug users alike, for non-medical use. Intentional or inadvertent tampering from extended release formulations will rapidly deliver a massive dose and produce profound pharmacologic effects.
[00617] Addicts and recreational drug users commonly use abusable drugs by a variety of routes of administration. Commonly used methods include 1) parenteral (e.g., intravenous injection, where the drug is crushed and extracted or melted and the contents of a dosage unit then injected), 2) intranasal (e.g., snorting, where the drug is inhaled as powdered dosage unit), and 3) episodic or repeated oral ingestion of crushed product, where the drug is chewed to increase the surface area and permit rapid release of drug substance. All of these strategies are intended to more efficiently get the abusable drug into the CNS, both in terms of total amount of drug, peak concentration of drug and time to peak concentration of drug.
[00618] One mode of abuse involves the extraction of the drug component from the dosage form by first mixing the tablet or capsule with a suitable solvent (e.g., water or alcohol), and then filtering and/or extracting the drug component from the mixture for intravenous injection. Another mode of abuse of extended release drugs involves dissolving the drug in water, alcohol or another "recreational solvent" to hasten its release and to ingest the contents orally, in order to provide. high peak concentrations and maximum euphoriant effects.
[00619] It is necessary to be able to measure resistance or deterrence to the likely routes of abuse in a meaningful and relevant way. The in vitro tests below are provided for illustration of some testing methods and are intended to be non-limiting examples. This invention therefore contemplates the use of test methods other than those specifically disclosed herein, including those which may hereafter become known to the art to be capable of performing the necessary functions.
Extraction with Alcohol on Whole Dosage Unit
[00620] Method: Place a whole dosage unit in 18 mL of 0.1N HCl in a 60 mL amber bottle and shake at 240 rpm on an orbital shaker for 30 min. After 30 min add 12 mL of ethanol (95-96%) to each bottle. Swirl by hand and remove a 1 mL sample from each bottle (To). Place the solutions back in the orbital shaker for further shaking at 240 rpm. Take 1 mL samples after 10, 20, 30, 40, 60 and 180 min of further shaking for each bottle. Analyze and graph the results on a linear scale of cumulative release (%) vs. time (min).
Extraction with Alcohol on a Crushed or Cut Dosage Unit
[00621] Extension of the above test. Method: Place a tablet (after crushing with a single crush with a spatula) or a capsule (cut in half) in 18 mL of 0.1N HCl in a 60 mL amber bottle and shake at 240 rpm on an orbital shaker for 30 min. Continue the test as in 1) above.
Extraction into Water
[00622] Method: Crush with a mortar and pestle and grind in 5 mL of water for
5 minutes. The resulting suspension is filtered through a 0.45 micron filter into a flask and diluted to 50 mL with water. Quantify drug concentration by HPLC.
Freeze and Crush
[00623] Method: Freeze the dosage unit in a domestic freezer for 24 hr, then grind with a mortar and pestle for five minutes. Sieve through a suitable sieve (ca βOOmicron) and, by weighing, measure the percentage passing the sieve.
Taste of Base Excipient Mix (organoleptic tesf)
[00624] Method: Chew a placebo mix for five minutes and rate the taste on a 0-
10 scale with 0 as bland to repulsive at 10. This method is relevant only to dosage units containing taste modifiers.
Extraction into Acid
[00625] Method: Crush with a mortar and pestle and heat to boiling in 5 mL of vinegar. The resulting suspension is filtered through a 0.45 micron filter into a flask and diluted to 50 mL with water. Quantify drug concentration by HPLC.
Application of Heat (melting temperature >50°C or 55°O
[00626] Method: Heat the squashed contents of a dosage unit on a hot plate until melted. Determine the temperature of melting and test whether the mix becomes sufficiently fluid to be drawn up into a syringe via a 1.2 mm needle then expelled.
EXAMPLES
[00627] Non-limiting examples for preparing the dosage form are set forth below.
Detailed Description of the Preferred Embodiments [00628] The following examples illustrate various aspects of the present invention. They are not to be construed to limit the claims in any manner whatsoever.
[00629] A wide variety of methods known in the art for the preparation of immediate release and controlled release dosage forms may be incorporated into the invention.
[00630] Other suitable abusable drugs as defined in this invention may also be prepared by modification of the examples herein and by use of material other than those specifically disclosed herein, including those which may hereafter become known to the art to be capable of performing the necessary functions. Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered and obvious to those skilled in the art are within the spirit and scope of the invention.
[00631] More than abusable drug may be included in the dosage form either in an immediate release or extended release form
100632] The ingredients used for the preparation of the dosage form may be modified depending on the selection, dose and desired duration of effect of the abusable drug and any included aversive agent, pharmacologic antagonist or other non-abusable therapeutic agent. In some embodiments, a change in the dose or amount of abusable drug does not require a change in amount of other ingredients. In other embodiments, a proportional change in the amount of other ingredients is required to maintain the desired properties. In yet other embodiments, a change in the dose or amount of abusable drug necessitates a change in the nature and/or amount of ingredients to provide the required characteristics of the abusable drug (e.g., immediate release, sustained release, duration of effect, rate and extent of absorption, therapeutic concentrations and effect, abuse deterrence properties, protection against alcohol dose dumping; and/or protection against significant changes in bioavailability due to fed or fasted states etc.)
[00633] Optionally, aversive agents may be included sleeted from the group comprising (i) laxatives; (ii) cutaneous vasodilators; (iii) headache producing agents; (iv) emetics, emetogenic and nausea producing compounds; (iv) bittering agents (v) mucosal, naso-mucosal, oro-mucosal, respiratory, tissue and gastrointestinal irritants; (vi) tissue staining, non-tissue staining and beverage staining dyes, lakes and colorants; (vii) fecal and urine discolorants; (viii) malodorous agents; (ix) opioid antagonists; and (x) and (x) benzodiazepine antagonists (e.g., flumazenil), and mixtures thereof.
[00634] Manufacturing methods described herein are utilized for the preparation of the abusable drugs as shown in the examples below. Variations to the methods may be employed, in some embodiments, depending on the specific chemical, physicochemical, pharmaceutical and pharmacologic properties of the abusable drug, excipients and their interaction and other factors. Compositions and methods of the present invention provide: (i) abuse deterrence; (ii) extended release; and/or (iii) protection against alcohol dose dumping; and/or (iv) protection against significant changes in bioavailability due to fed or fasted states; and/or (v) simultaneously providing more than one of foregoing (i) to (iv); wherein the dosage form is prepared using compounds selected from the group consisting of: (a) hydrogenated Type I or Type II vegetable oils; (b) polyoxyethylene stearates and distearates; (c) glycerol monostearate; (d) poorly water soluble, high melting point (mp = 45 to 100° C) waxes, and mixtures thereof.
[00635] As shown in further examples below, any abusable drug of the invention may be prepared to provide (i) abuse deterrence; or (ii) extended release; or (iii) resistance against alcohol dose dumping; or (iv) resistance against alcohol dose dumping when formulated as extended release; or (v) protection against significant changes in bioavailability due to fed or fasted states; or (vi) more than one of the foregoing [(i) to (v)] properties; said invention comprising one or more abusable drugs and one or more ADER agents; said invention, in some embodiments, providing further abuse deterrence through the use of aversive agents; said invention, in some embodiments, using substantially the same ADER agents to achieve the foregoing (i) to (v); said invention, in some embodiments, providing more than one of the foregoing [(i) to (v)] properties; said invention, in some embodiments, comprising one or more abusable drugs and at least two ADER agents; said invention, in some embodiments, comprising at least two ADER agents selected from the group consisting of: (a) hydrogenated Type I or Type II vegetable oils; (b) polyoxyethylene stearates and distearates; (c) glycerol monostearate; (d) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said invention, in some embodiments, comprising at least two ADER agents selected from at least two categories [(a) to (d)] from the group consisting of: (a) hydrogenated Type I or Type II vegetable oils; (b) polyoxyethylene stearates and distearates; (c) glycerol monostearate; (d) poorly water soluble, high melting point (mp — 45 to 100° C) waxes; said invention, in some embodiments, further comprising an immediate release form of the abusable drug; said invention, in some embodiments, further comprising an immediate release form of the abusable drug in solution; said invention, in some embodiments, simultaneously providing more than one of foregoing (i) to (v); said invention, in some embodiments, simultaneously providing more than one of foregoing (i) to (v) using substantially the same ADER agents.
[00636] Example 1 to 75 may be prepared as follows: (i) Dispense the hydrogenated Type I vegetable oil, hydrogenated Type II vegetable oil, polyoxyethylene stearates, polyoxyethylene distearates, glycerol monostearate and/or poorly water soluble, high melting point (mp = 45 to 100° C) waxes into a mixer; (ii) Heat until fully melted; (iii) dispense the hydroxypropyl methyl cellulose (HPMC) into the mixer; (iv) Mix until dispersed; (v) Dispense the Aerosil into the same vessel; (vi) Mix until dispersed; (vii) Dispense the desired abusable drug or mixture of abusable drugs into the same vessel; (viii) Stir thoroughly with a high shear mixer; (ix) Transfer the mix into a liquid filling machine; (x) Fill into hard gelatin (or HPMC) capsule; (xi) Optionally, transfer the capsules to a banding machine and band the capsules.
EXAMPLE 1 [006371
Figure imgf000194_0001
[00638]
[00639]
[00640]
Figure imgf000195_0001
[00641]
[00642]
Figure imgf000196_0001
EXAMPLE 7
[00643]
Figure imgf000196_0002
EXAMPLE 8
[00644] [00645]
[00646]
[00647]
Figure imgf000197_0001
EXAMPLE 12
[00648] [00649]
Figure imgf000198_0001
EXAMPLE 14
[00650]
[00651]
Figure imgf000198_0002
[00652]
[00653]
[00654]
Figure imgf000199_0001
[00655]
[00656]
Figure imgf000200_0001
EXAMPLE 21
[00657]
Figure imgf000200_0002
[00658]
Figure imgf000201_0001
EXAMPLE 23
[00659]
[00660]
Figure imgf000201_0002
EXAMPLE 25
(00661]
[00662]
[00663]
[00664]
Figure imgf000202_0001
[00665]
[00666]
[00667]
[00668]
Figure imgf000203_0001
[00669]
[00670]
[00671]
[00672]
Figure imgf000204_0001
[00673]
[00674]
[00675]
[00676]
Figure imgf000205_0001
[00677] EXAMPLE 41
[00678]
[00679]
[00680]
Figure imgf000206_0001
[00681] EXAMPLE 45
[00682]
100683]
[00684]
[00685]
Figure imgf000207_0001
[00686]
[00687]
[00688]
[00689]
Figure imgf000208_0001
[00690]
[00691]
[00692]
[00693]
Figure imgf000209_0001
[00694]
[00695]
[00696]
[00697]
Figure imgf000210_0001
[00698] EXAMPLE 62
[00699]
[00700]
[00701]
Figure imgf000211_0001
[00702]
[00703]
[00704]
Figure imgf000212_0001
EXAMPLE 69
[00705]
Figure imgf000213_0001
EXAMPLE 70
[00706]
[00707]
[00708]
Figure imgf000213_0002
[00709]
[00710]
[00711]
Figure imgf000214_0001
Figure imgf000215_0001
[00712] Example 16 to 150 may be prepared as using the formula of Examples
1 to 75, respectively with the following modification: (i) Dispense the hydrogenated Type I vegetable oil, hydrogenated Type II vegetable oil, polyoxyethylene stearates, polyoxyethylene distearates, glycerol monostearate and/or poorly water soluble, high melting point (mp = 45 to 100° C) waxes into a mixer; (ii) Heat until fully melted, (iii) dispense the hydroxypropyl methyl cellulose (HPMC) into the mixer; (iv) Mix until dispersed; (v) Dispense the Aerosil into the same vessel; (vi) Mix until dispersed; (vii) Dispense the desired abusable drug or mixture of abusable drugs into the same vessel; (viii) Stir thoroughly with a high shear mixer; (ix) Feed the blended material continuously into a twin screw extruder and collect the resultant strands on a conveyor and allow to cool (the conveyor is set to provide extrudate diameter of 0.5 ram or 1 mm and the pelletizer is set to provide pellets of approximately 0.5 or 1 mm length); (x) Screen the pellets so formed through a sieve and collect the desired particle size (sieve portion); (xi) Fill pellets into capsules or blend the pellets with talc and magnesium stearate and compress into tablets.
[00713] These and other embodiments of the present invention will readily occur to those of ordinary skill in the art in view of the disclosure herein.
[00714] The included examples are illustrative but not limiting of the methods and composition of the present invention. Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered and obvious to those skilled in the art are within the spirit and scope of the invention.
[00715] A wide variety of materials can be used for preparing the dosage form according to this invention. This invention therefore contemplates the use of materials other than those specifically disclosed herein, including those which may hereafter become known to the art to be capable of performing the necessary functions. Having now fully described the invention, it will be understood to those of ordinary skill in the art that the same can be performed within a wide and equivalent range of conditions, formulations, and other parameters without affecting the scope of the invention or any embodiment thereof. All patents and publications cited herein are fully incorporated by reference herein in their entirety.

Claims

WHAT IS CLAIMED IS:
1. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds rendering said dosage form abuse resistant.
2. The oral dosage form of claim 1, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant.
3. The oral dosage form of claim 1, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant.
4. The oral dosage form of claim 1, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant.
5. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds rendering said dosage form extended release.
6. The oral dosage form of claim 5, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds rendering said dosage form extended release.
7. The oral dosage form of claim 5, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds rendering said dosage form extended release.
8. The oral dosage form of claim 5, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds rendering said dosage form extended release.
9. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds rendering said dosage form abuse resistant and extended release.
10. The oral dosage form of claim 9, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant and extended release.
11. The oral dosage form of claim 9, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant and extended release.
12. The oral dosage form of claim 9, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant and extended release.
13. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds rendering said dosage form abuse resistant and extended release using substantially the same ingredients.
14. The oral dosage form of claim 13, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant and extended release using substantially the same ingredients.
15. The oral dosage form of claim 13, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant and extended release using substantially the same ingredients.
16. The oral dosage form of claim 13, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds rendering said dosage form abuse resistant and extended release using substantially the same ingredients.
17. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp — 45 to 100° C) waxes; said compounds rendering said dosage form resistant to dose dumping due upon co-ingestion intact with alcohol.
18. The oral dosage form of claim 17, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds rendering said dosage form resistant to dose dumping due upon co- ingestion intact with alcohol.
19. The oral dosage form of claim 17, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds rendering said dosage form resistant to dose dumping due upon co-ingestion intact with alcohol.
20. The oral dosage form of claim 17, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds rendering said dosage form resistant to dose dumping due upon co-ingestion intact with alcohol.
21. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds rendering said dosage form resistant to significant changes in oral bioavailability due to changes in food intake (i.e., fed or fasted states).
22. The oral dosage form of claim 21, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds rendering said dosage form resistant to significant changes in oral bioavailability due to changes in food intake (i.e., fed or fasted states).
23. The oral dosage form of claim 21, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds rendering said dosage form resistant to significant changes in oral bioavailability due to changes in food intake (i.e., fed or fasted states).
24. The oral dosage form of claim 21, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds rendering said dosage form resistant to significant changes in oral bioavailability due to changes in food intake (i.e., fed or fasted states).
25. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds rendering said dosage form floatable or buoyant; said compounds preventing said dosage form from rapidly sinking; said compounds rendering said dosage form resistant to intentional or surreptitious adulteration of beverages.
26. The oral dosage form of claim 25, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds rendering said dosage form floatable or buoyant; said compounds preventing said dosage form from rapidly sinking; said compounds rendering said dosage form resistant to intentional or surreptitious adulteration of beverages.
27. The oral dosage form of claim 25, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds rendering said dosage form floatable or buoyant; said compounds preventing said dosage form from rapidly sinking; said compounds rendering said dosage form resistant to intentional or surreptitious adulteration of beverages.
28. The oral dosage form of claim 25, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds rendering said dosage form floatable or buoyant; said compounds preventing said dosage form from rapidly sinking; said compounds rendering said dosage form resistant to intentional or surreptitious adulteration of beverages.
29. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds simultaneously rendering said dosage form: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; (e) resistant to intentional or surreptitious adulteration of beverages; or (f) two or more of the foregoing [(a) to (e)].
30. The oral dosage form of claim 29, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds simultaneously rendering said dosage form: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; (e) resistant to intentional or surreptitious adulteration of beverages; or (f) two or more of the foregoing [(a) to (e)].
31. The oral dosage form of claim 29, wherein two or more compounds selected are from at least two said ' categories [(i) to (iv)]; said compounds simultaneously rendering said dosage form: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; (e) resistant to intentional or surreptitious adulteration of beverages; or (f) two or more of the foregoing [(a) to
(e)].
32. The oral dosage form of claim 29, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds simultaneously rendering said dosage form: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; (e) resistant to intentional or surreptitious adulteration of beverages; or (f) two or more of the foregoing [(a) to
(e)].
33. The oral dosage form of any of claims 29 to 32, said compounds simultaneously rendering said dosage form with three or more of the properties selected from the group comprising: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; (e) resistant to intentional or surreptitious adulteration of beverages.
34. The oral dosage form of any of claims 29 to 33, wherein said compounds render said dosage form with two or more or three or more of the properties selected from the group comprising: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and/or (e) resistant to intentional or surreptitious adulteration of beverages; using substantially the same ingredients.
35. An oral dosage form comprising: (a) an abusable drug or a pharmaceutically acceptable salt thereof or a mixture thereof and (b) one or more compounds selected from the categories [(i) to (iv)] from the group consisting of: (i) hydrogenated Type I or Type II vegetable oils; (ii) polyoxyethylene stearates and distearates; (iii) glycerol monostearate; (iv) poorly water soluble, high melting point (mp = 45 to 100° C) waxes; said compounds reducing or preventing toxicity from intentional or inadvertent tampering of the dosage form.
36. The oral dosage form of claim 35, wherein two or more compounds selected are from said categories [(i) to (iv)]; said compounds reducing or preventing toxicity from intentional or inadvertent tampering of the dosage form.
37. The oral dosage form of claim 35, wherein two or more compounds selected are from at least two said categories [(i) to (iv)]; said compounds reducing or preventing toxicity from intentional or inadvertent tampering of the dosage form.
38. The oral dosage form of claim 35, wherein three or more compounds selected are from at least three said categories [(i) to (iv)]; said compounds reducing or preventing toxicity from intentional or inadvertent tampering of the dosage form.
39. A method of rendering an abusable drug: (a) abuse resistant; (b) extended release; (c) resistant to dose dumping due to alcohol; (d) resistant to significant changes in oral bioavailability due to changes in food intake; and/or (e) resistant to intentional or surreptitious adulteration of beverages comprising administering to a human patient in need thereof an effective amount of the oral dosage form of any of claims 1-38.
40. A claim according to any of claims 1 to 39, wherein the abusable drug is a compound selected from the group comprising amphetamine, amphetamine like CNS-stimulants, amphetamine analogs, CNS- stimulants, nicotine, methylphenidate, modafinil, alkylxanthines, anorectics, psychostimulants, benzodiazepine agonists, CNS depressants, barbiturates, cannabinoid agonists, anxiolytic agents, sedatives, non-benzodiazepine hypnotics, psychoactive agents, psychostimulant, controlled substances (i.e., scheduled drugs under the United States Controlled Substances Act of 1970, as amended) and drugs listed under the United States Psychotropic Substances Act of 1978, as amended and prodrugs, analogs, derivatives, pharmaceutically acceptable salts thereof and mixtures thereof in racemic or enantiomeric form, excluding opioid agonists.
41. A claim according to claim 40, wherein the abusable drug is a benzodiazepine agonist selected from the group consisting of alprazolam, bromazepam, brotizolam, camazepam, chlordiazepoxide, cinolazepam, clobazam, clonazepam, clorazepate, desalkylflurazepam, diazepam, estazolam, fiunitrazepam, flurazepam, halazepam, ketazolam, loprazolam, lorazepam, lormetazepam, medazepam, metaclazepam, midazolam, nitrazepam, nordazepam, oxazepam, phenazepam, pinazepam, prazepam, quazepam, temazepam, tetrazepam, triazolam, zaleplone, Zolpidem and zopiclone, and prodrugs, analogs, derivatives, pharmaceutically acceptable salts thereof and mixtures thereof in racemic or enantiomeric form.
42. A claim according to claim 40, wherein the abusable drug is a non- benzodiazepine hypnotics and CNS depressants selected from the group comprising sodium oxybate, diphenhydramine, chlorpheniramine, trazadone, amitriptyline, cyclobenzaprine, methocarbamol, carisoprodol and ramelteon, and prodrugs, analogs, derivatives, pharmaceutically acceptable salts thereof and mixtures thereof in racemic or enantiomeric form.
43. A claim according to claim 40, wherein the abusable drug is a cannabinoid agonists selected from the group comprising dexanabinol (HU211), BAY 38-7271, Naphthalen-l-yl-(4-pentyloxynaphthalen-l- yl)methanone, THC (delta-9-tetrahydrocannabinol), nabilone, dronabinol, cannabidiol, cannabinol, cannabichromene, cannabigerol, cannabigerol, anandamide, (R)-methanandamide, 2- arachidonoylglycerol, HU210, desacetyllevonantradol, CP55940, CP55244, URB602, or WIN55212-2 and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof.
44. A claim according to claim 40, wherein the abusable drug is a cannabinoid agonists selected from the group comprising 9-THC propyl analog, endocannabinoids, cannabinoid terpenoids, cannabinoid flavonoids, inhibitors of cannabinoid agonist metabolism, inhibitors of monoacylglycerol lipase, cannabidiol propyl analogues, cannabichromene propyl analogues, THC-like ABC tricyclic cannabinoid analogues, synthetic AC bicyclic cannabinoid analogues, synthetic ACD tricyclic cannabinoid analogues, aminoalkylindole compounds or analogs of 2-Arylimino-5,6-dihydro-4H-l,3-thiazines and their or their pharmaceutically acceptable salts, prodrugs, esters, analogs, derivatives, solvates, complexes, polymorphs, hydrates and metabolites, as racemates or an individual diastereoisomers or enantiomeric isomers thereof or mixtures thereof.
45. A claim according to claim 40, wherein the abusable drug is a CNS- stimulants, psychostimulants, alkylxanthine, or anorectic compound selected from the group comprising adrafanil, alkyxanthine derivatives, almitrine, amfetaminil, aminophylline, amiphenazole, ammonium camphocarbpnate, amphetamine, bamifylline, benzfetamine, brolamfetamine, caffeine, cathine [(+)-norpseudoephedrine], cathinone, celastrin, chlorphentermine, clonobenzorex, cropropamide, crotetamide, deanol, dextroamphetamine, diethylaminoethanol, diethylpropion, dimfline, doxapram, doxofylline, diprophylline, dyphylline, etamivan, etofylline, enprophylline, etamiphylline, methylphenidate, dexmethylphenidate, fencamfamin, fenetylline, fenozolone, fenproporex, lisdexamfetamine, Hsdexamfetamine dimesylate, lisofylline, lobeline, mazindol, mefenorex, mepixanox, methamphetamine, methylenedioxymethamphetamine, modafinil, nicotine, nikethamide, oxtriphylline, pemoline, pentetrazol, phedimetrazine, phenmetrazine, phentermine, pentoxifylline, phenylpropanolamine, pipradrol, prethcamide, prolintane, propylhexedrine, propentofylline, pentifylline, pseudoephedrine, pyridophylline, proxyphylline, sibutramine, tenamfetamine (methylenedioxeamphetamine), theophylline and theobromine, and prodrugs, analogs, derivatives, pharmaceutically acceptable salts thereof and mixtures thereof in racemic or enantiomeric form.
46. A claim according to claim 1 to 40, wherein the dosage form is in immediate release form instead of extended release form.
47. A claim according to claim 1 to 40, further comprising one or more or two or more aversive agent selected from the group comprising: (i) antagonists of abusable drugs; (ii) laxatives; (iii) cutaneous vasodilators; (iv) headache producing agents; (v) emetics, emetogenic and nausea producing compounds; (vi) bittering agents (vii) mucosal, naso-mucosal, oro-mucosal, respiratory, tissue and gastrointestinal irritants; (viii) drugs that precipitate pharmacologic withdrawal effects; (ix) tissue staining, non-tissue staining and beverage staining dyes, lakes and colorants; (x) fecal and urine discolorants; (xi) malodorous agents; and (xii) pharmacologic antagonists to co-abused drugs, said co-abused drugs not part of the dosage form.
48. A claim according to claim 47, wherein the aversive agent is a bittering agent selected from the group comprising T2R or TAS2R receptor agonists, phenylthiourea (phenylthiocarbamide), natural, artificial and synthetic flavor oils, flavoring aromatics, flavoring oils, oleoresins, speaπnint oil, peppermint oil, eucalyptus oil, oil of nutmeg, allspice, mace, oil of bitter almonds, menthol, citrus oils including lemon, orange, lime, grapefruit, and fruit essences, sucrose derivatives, sucrose octaacetate, chlorosucrose derivatives, quinine, denatonium, denatonium saccharide and denatonium benzoate.
49. A claim according to claim 47, wherein the aversive agent is a naso- mucosal, oro-mucosal, respiratory or tissue irritants selected from the group comprising transient receptor potential vanilloid 1 agonists, resiniferanoids, capsaicinoids, phorboid vanilloids, terpenoid 1,4- unsaturated dialdehydes, capsaicin, capsaicin analogs, resiniferatoxin, olvanil, piperine, zingerone, anandamide, 12- and 15-(S)-hydroperoxy- eicosatetraenoic acids, 5 and 15-(S)-hydroxyeicosatetraenoic acids, phorbol 12-phenylacetate 13-acetate 20-homovanillate, 2 phorbol 12,13-didecanoate 20-homovanillate, leukotriene B(4), tinyatoxin, heptanoylisobutylamide, N-(3-acyloxy-2-benzylpropyl)-N'- dihydroxytetrahydrobenzazepine, tetrahydroisoquinoline thiourea analogs, heptanoyl guaiacylamide, isobutylamides, guaiacylamides, dihydrocapsaicin, homovanillyl octylester, nonanoyl vanillylamide, formic acid, acetic acid, propionic acidy, butyric acid, valeric acid, caproic acid, caprillic acid, capric acid, oxalic acid, malonic acid, succicnic acid, glutaric acid, adipic acid, maleic acid, fumaric acid, citric acid, sodium lauryl sulfate, poloxamer, sorbitan mόnoesters, glyceryl monooleates, niacin, mustard, allyl isothiocyaanate and p- hydroxybenzyl isothiocyanate and acetylsalicylic acid.
50. A claim according to claim 47, wherein the aversive agent is an emetogenic or nausea producing agents selected from the group comprising zinc and pharmaceutically acceptable salts thereof, dopamine agonists, apomorphine, ipecac, ipecacuanha, emetine, methylcephaeline, cephaeline, psychotrine, O-methylpsychotrine, ammonium chloride, potassium chloride, magnesium sulfate, ferrous gluconate, ferrous sulfate, aloin, algarot or antimonious oxychloride, antimony trichloride, folate, folic acid, niacin and nicotinamide.
51. A claim according to claim 47, wherein the aversive agent is a cutaneous vasodilator selected from trie group comprising niacin, nicotimiric acid, beta-hydroxybutyrate and nicotinic receptor agonists, including agonists at nicotinic receptor HM74A and nicotinic receptor GPRl 09 A.
52. A claim according to any claim of claim 1 to 51, said dosage form providing a therapeutic effect for at least about 6, 8, 12 or 24 hours.
53. A claim according to any claim of claim 1 to 39, wherein the hydrogenated vegetable oil is selected from the group comprising hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenated soybean oil and hydrogenated palm kernel oil.
54. A claim according to any claim of claim 1 to 39, wherein the polyoxyethylene stearate and distearates is selected from the group comprising polyoxyl 2, 4, 6, 8, 12, 20, 30, 40, 50, 100 and 150 stearates and polyoxyl 4, 8, 12, 32 and 150 distearates.
55. A claim according to any claim of claim 1 to 39, wherein the poorly water soluble, high melting point (mp = 45 to 100° C) wax is selected from the group comprising animal waxes, insect waxes, vegetable waxes, mineral waxes, petroleum waxes, synthetic waxes, nonionic emulsifying waxes, anionic emulsifying wax, carnauba wax, microcrystalline wax, yellow wax, white wax, cetyl esters wax, hydrogenated castor oil, lanolin alcohols, lanolin, glyceryl palmitostearate, cetostearyl alcohol and beeswax.
56. A claim according to any claim of claim 1 to 39, wherein the dosage form includes cellulose and cellulose derivatives selected from the group comprising cellulose acetate, microcrystalline cellulose, powdered cellulose, cellulose acetate phthalate, hydroxyethyl cellulose, silicified microcrystalline cellulose, hydroxypropyl cellulose, hydroxyethylmethyl cellulose, low-substituted hydroxypropyl cellulose, carboxymethylcellulose, carboxymethylcellulose calcium, hypromellose acetate succinate, hypromellose phthalate and ethylcellulose.
57. A claim according to any claim of claim 1 to 39, wherein the dosage form includes coconut oil products and derivatives, chosen from the group comprising coconut oil, hydrogenated coconut oil, fractionated coconut oil, cetyl alcohol, lauric acid and medium chain triglycerides.
58. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and said dosage form after administration to a human patient provides a mean ratio of the time to confirmed perceptible pain relief after administration of the intact dosage form to the time to confirmed perceptible pain relief after administration of the tampered dosage form of less than 10:1.
59. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and said dosage form after administration to a human patient provides a mean ratio of the time to meaningful pain relief after administration of the intact dosage form to the time to meaningful pain relief after administration of the tampered dosage form of less than 10:1.
60. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and said dosage form after administration to a human, patient provides a mean ratio of the peak pain intensity difference score after administration of the tampered dosage form to the peak pain intensity difference score after administration of the intact dosage form
J of less than 10:1.
61. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and said dosage form after administration to a human patient provides a mean ratio of the peak pain relief score after administration of the tampered dosage form to the peak pain relief score after administration of the intact dosage form of less than 10:1.
62. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and said dosage form after administration to a human patient provides a mean ratio of change from baseline to two hours post-dose in pain intensity score after administration of the tampered dosage form to the change from baseline to two hours post-dose in pain intensity score after administration of the intact dosage form of less than 10:1.
63. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and said dosage form after administration to a human patient provides a mean ratio of the number of patients with pain who need to be treated to obtain > 50% pain relief in one patient (i.e., number needed to treat or ISfNT) after administration of the tampered dosage form to the NNT after administration of the intact dosage form of less than 15:1, said NNT measured 0.5 to 4 hours after administration of the dosage form.
64. A claim according to any of claims 1 to 39, wherein said abusable drug is a sedating analgesic and said dosage form after administration to healthy subjects (who are naive to said analgesic) provides a mean ratio of the number needed to harm (NNH) due to moderate or severe sedation or drowsiness after administration of the tampered dosage form to the number needed to harm (NNH) due to moderate or severe sedation or drowsiness after administration of the intact dosage form of less than 10:1, said NNH measured 0.5 to 4 hours after administration of the dosage form.
65. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and said dosage form after administration to healthy subjects who are naϊve to said abusable drug and who are occasional or light consumers of alcohol, provides a mean ratio of the number needed to harm (NNH) due to moderate or severe sedation or drowsiness after administration of the tampered dosage form to the number needed to harm (NNH) due to moderate or severe sedation or drowsiness after administration of the intact dosage form of less than 10:1, said NNH measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
66. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and the mean ratio of the drug liking score in drug abusers and recreational drug users without pain after administration of the tampered dosage form to the drug liking score after administration of the of the intact dosage form is less than 10:1, said drug liking score measured 0.5 to 4 hours after administration of the dosage form.
67. A claim according to any of claims 1 to 39, wherein said abusable drug is an analgesic and the mean ratio of the drug effect score in drug abusers and recreational drug users without pain after administration of the tampered dosage form to the drug effect score after administration of the intact dosage form is less than 10:1, said drug effect score measured 0.5 to 4 hours after administration of the dosage form.
68. A claim according to any of claims 1 to 39, wherein the patient has a medical condition amenable to treatment with said abusable drug and the mean ratio of the NNT to obtain > 50% reduction in the cardinal sign or symptom of said medical condition after administration of the tampered dosage form to the NNT after administration of the intact dosage form is less than 10:1, said NNT measured 0.5 to 4 hours after administration of the dosage form.
69. A claim according to any of claims 1 to 39, wherein said abusable drug is a CNS stimulant or psychostimulant and said dosage form after administration to healthy subjects who are naϊve to said abusable drug, wherein the mean ratio of the NNH due to the incidence of tachycardia (i.e., resting heart rate > 100 beats per minute) after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 10:1, said NNH measured 0.5 to 4 hours after administration of the dosage form.
70. A claim according to any of claims 1 to 39, wherein said abusable drug is a CNS stimulant or psychostimulant and the mean ratio of the NNH due to the time to onset of sleep in healthy subjects who are naϊve to said abusable drug, after administration of the tampered dosage form, to the NNH after administration of the intact dosage form is less than 10:1.
71. A claim according to any of claims 1 to 39, where upon administration of abusable drug dosage form to drug abusers and recreational drug users, the mean ratio of the drug liking score after administration of the tampered dosage form to the drug liking score after administration of the intact dosage form is less than 10:1, said drug liking score measured 0.5 to 6 hours after administration of the dosage form.
72. A claim according to any of claims 1 to 39, where upon administration of abusable drug dosage form to drug abusers and recreational drug users, the mean ratio of the drug effect score after administration of the tampered dosage form to the drug effect score after administration of the intact dosage form is less than 10:1, said drug effect score measured 0.5 to 6 hours after administration of the dosage form.
73. A claim according to any of claims 1 to 39, where upon administration of abusable drug dosage form to drug abusers and recreational drug users, the mean ratio of the score on the "take again" questionnaire after administration of the tampered dosage form to the "take again" score after administration of the intact dosage form is less than 10:1, said "take again" score measured 0.5 to 6 hours after administration of the dosage form.
74. A claim according to any of claims 1 to 39, where upon administration of abusable drug dosage form to drug abusers and recreational drug users, the mean ratio of the score on the "coasting" questionnaire after administration of the tampered dosage form to the "coasting" score after administration of the intact dosage form is less than 10:1, said "coasting" score measured 0.5 to 6 hours after administration of the dosage form.
75. A claim according to any of claims 1 to 39, wherein said abusable drug is a CNS depressant or produces sedation or drowsiness and the mean ratio of the impairment on the "critical tracking task" driving skills test score after administration to healthy subjects who are naϊve to said abusable drug, of the tampered dosage form, to the "critical tracking task" driving skills test score after administration of the intact dosage form is less than 10:1, said "critical tracking task" driving skills test score measured 0.5 to 6 hours after administration of the dosage form.
76. A claim according to any of claims 1 to 39, wherein said abusable drug is a CNS depressant or produces sedation or drowsiness and the mean ratio of the impairment on the "stop signal task" driving skills test score after administration to healthy subjects who are naive to said abusable drug, of the tampered dosage form, to the "stop signal task" driving skills test score after administration of the intact dosage form is less than 10:1, said ""stop signal task" driving skills test score measured 0.5 to 6 hours after administration of the dosage form.
77. A claim according to any of claims 1 to 39, wherein said abusable drug is a CNS depressant or produces sedation or drowsiness and the mean ratio of the impairment on the "Tower of London" driving skills test score after administration to healthy subjects who are naive to said abusable drug, of the tampered dosage form, to the "Tower of London" driving skills test score after administration of the intact dosage form is less than 10:1, said "Tower of London" driving skills test score measured 0.5 to 6 hours after administration of the dosage form.
78. A claim according to any of claims 1 to 39, wherein the patient has a medical condition amenable to treatment with said abusable drug and the mean ratio of the peak relief score on the cardinal sign or symptom of said medical condition after administration of the tampered dosage form to the peak relief score after administration of the intact dosage form is less than 10:1.
79. A claim according to any of claims 1 to 39, wherein the patient has a medical condition amenable to treatment with said abusable drug and the mean ratio of change from baseline to two hours post-dose in score on the cardinal sign or symptom of said medical condition after administration of the tampered dosage form to the change from baseline to two hours post-dose in score after administration of the intact dosage form is less than 10:1.
80. A claim according to any of claims 1 to 39, wherein said abusable drug in the dosage form is a CNS depressant or produces sedation or drowsiness, wherein the mean ratio of the NNH due to moderate or severe sedation or drowsiness in healthy subjects, who are naϊve to said abusable drug and who are occasional or light consumers of alcohol, after administration of the tampered dosage form to the NNH after administration of the intact dosage form is less than 10:1, said NNH measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
81. A claim according to any of claims 1 to 39, wherein said abusable drug in the dosage form is a CNS depressant or produces sedation or drowsiness, wherein the mean ratio of the impairment on the "critical tracking task" driving skills test score in healthy subjects, who are naϊve to said abusable drug and who are occasional or light consumers of alcohol, after administration of the tampered dosage form to the "critical tracking task" driving skills test score after administration of the intact dosage form is less than 10:1, said "critical tracking task" driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
82. A claim according to any of claims 1 to 39, wherein said abusable drug in the dosage form is a CNS depressant or produces sedation or drowsiness, wherein the mean ratio of the impairment on the "stop signal task" driving skills test score in healthy subjects, who are naϊve to said abusable drug and who are occasional or light consumers of alcohol, after administration of the tampered dosage form to the "stop signal task" driving skills test score after administration of the intact dosage form is less than 10:1, said "stop signal task" driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
83. A claim according to any of claims 1 to 39, wherein said abusable drug in the dosage form is a CNS depressant or produces sedation or drowsiness, wherein the mean ratio of the impairment on the "Tower of London" driving skills test score in healthy subjects, who are naive to said abusable drug and who are occasional or light consumers of alcohol, after administration of the tampered dosage form to the "Tower of London" driving skills test score after administration of the intact dosage form is less than 10:1, said "Tower of London" driving skills test score measured 2.5 to 6 hours after administration of the dosage form, said dosage form administration followed about 1.5 hours later by alcohol (ethanol) administration sufficient to maintain a blood alcohol concentration of 0.04% to 0.08%.
84. A claim according to any of claims 1 to 39 providing an in-vitro release rate by weight of an abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a
• pH of between 1.6 and 7.2 at 37 0C of between 0% and about 60% at 1 hour, between about 0% and about 80% at 2 hours, between about 3% and about 95% at 4 hours and between about 10% and about 100% at 8 hours.
85. A claim according to any of claims 1 to 39 providing an in-vitro release rate by weight of an abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% to about 47.5% at 1 hour, from about 10% to about 65% at 2 hours, from about 15% to about 70% at 4 hours, from about 25% to about 77.5% at 6 hours, from about 35% to about 87.5% at 9 hours, and greater than about 65% at 12 hours.
86. A claim according to any of claims 1 to 39 providing an in-vitro release rate by weight of an abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 5% and about 50% at 1 hour, between about 10% and about 75% at 2 hours, between about 20% and about 95% at 4 hours, between about 40% and about 100% at 8 hours, greater than about 50% at 12 hours, greater than about 70% at 18 hours, and greater than about 80% at 24 hours.
87. A claim according to any of claims 1 to 39 providing an in-vitro release rate by weight of an abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 1% and about 45% at 1 hour, between about 5%, and about 7s0% at 2 hours, between about 10% and about 90% at 4 hours, between about 20% and about 90% at 8 hours, greater than about 60% at 12 hours, greater than about 80% at 18 hours, and greater than about 85% at 24 hours.
88. A claim according to any of claims 1 to 39 providing an in-vitro release rate by weight of an abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 5% and about 60% at 1 hour, between about 12.5% and about 80% at 2 hours, between about 25% and about 95% at 4 hours, between about 45% and about 100% at 8 hours, greater than about 55% at 12 hours, greater than about 65% at 18 hours, and greater than about 70% at 24 hours.
89. A claim according to any of claims 1 to 39 providing an in-vitro release rate by weight of an abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% to about 30% at 1 hour, from about 10% to about 65% at 4 hours, from about 20% to about 70% at 8 hours, from about 25% to about 80% at 12 hours, from about 35% to about 95% at 18 hours, and greater than about 65% at 24 hours.
90. A claim according to any of claims 1 to 39 providing an in-vitro release rate by weight of an abusable drug, when measured by the USP Basket and Paddle Methods at 100 rpm in 900 mL aqueous buffer at a pH of between 1.6 and 7.2 at 37 0C of between 0% and about 50% at 1 hour, between about 0% and about 75% at 2 hours, between about 3% and about 95% at 4 hours, between about 10% and about 100% at 8 hours, between about 25% and about 100% at 12 hours, between about 30% and about 100% at 16 hours, between about 50% and about 100% at 24 hours, and greater than about 80% at 36 hours.
91. A claim according to any of claims 1 to 39, wherein the dosage form is bioequivalent when taken under with and without 120 mL of a 40% ethanol solution or with and without 240 mL of a 40% ethanol solution.
92. A claim according to any of claims 1 to 39, wherein the dosage form upon administration provides a mean fed to fasted abusable drug AUCo-∞ difference of less than 35%, or less than 25%, or less than 15%, or less than 10%.
93. A claim according to any of claims 1 to 39, wherein the dosage form upon administration provides a mean fed to fasted abusable drug Cmax difference of less than 35%, or less than 25%, or less than 15%, or less thanlθ%.
94. A claim according to any of claims 1 to 39, wherein the dosage form upon administration provides a mean alcohol state to no alcohol state abusable drug Cmax difference of less than 35%, or less than 25%, or less than 15%, or less thanlθ%.
95. A claim according to any of claims 1 to 39 wherein the mean ratio of the amount of abusable drug released from the dosage form after mechanical tampering to the amount of abusable drug released from the intact dosage form based on the dissolution at 0.5 hours of the dosage form in 900 mL of Simulated Gastric Fluid using the USP Basket and Paddle Methods at 50 rpm at 37 degrees 0C is less than 10:1.
96. A method for treating or preventing medical conditions amenable to treatment with abusable drugs comprising administering to a human patient in need thereof an effective amount of the oral dosage form of any of claims 1-95.
97. A method for treating or preventing pain, comprising administering to a human patient in need thereof an effective amount of the oral dosage form of any of claims 1 -95.
PCT/US2007/019745 2006-09-11 2007-09-12 Multimodal abuse resistant and extended release formulations WO2008033351A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US84339206P 2006-09-11 2006-09-11
US60/843,392 2006-09-11
US90798707P 2007-04-26 2007-04-26
US60/907,987 2007-04-26

Publications (2)

Publication Number Publication Date
WO2008033351A2 true WO2008033351A2 (en) 2008-03-20
WO2008033351A3 WO2008033351A3 (en) 2009-04-09

Family

ID=39184298

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/019745 WO2008033351A2 (en) 2006-09-11 2007-09-12 Multimodal abuse resistant and extended release formulations

Country Status (1)

Country Link
WO (1) WO2008033351A2 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009112436A1 (en) * 2008-03-10 2009-09-17 Eurodrug Laboratories B.V. Modified release composition comprising doxofylline
US20120100212A1 (en) * 2009-07-06 2012-04-26 Kyorin Pharmaceutical, Co., Ltd. Tablet having hollow structure
US8329744B2 (en) 2005-11-02 2012-12-11 Relmada Therapeutics, Inc. Methods of preventing the serotonin syndrome and compositions for use thereof
AT509000B1 (en) * 2009-10-23 2012-12-15 Rausch Peter WATER-SOLUBLE PREPARATIONS OF CANNABINOIDS AND CANNABIC PREPARATIONS AND THEIR APPLICATIONS
US20130098286A1 (en) * 2010-07-06 2013-04-25 Catherine Herry Method and agent for detecting drugs in beverages
US20130108556A1 (en) * 2010-07-06 2013-05-02 Ethypharm Pharmaceutical form for combating chemical submission of a medicament
US8920838B2 (en) 2006-08-03 2014-12-30 Horizon Pharma Ag Delayed-release glucocorticoid treatment of rheumatoid disease
US9125833B2 (en) 2005-11-02 2015-09-08 Relmada Therapeutics, Inc. Multimodal abuse resistant and extended release opioid formulations
US9125867B2 (en) 2010-02-24 2015-09-08 Invincible Biotechnology Diversion- and/or abuse-resistant compositions and methods for making the same
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US10159268B2 (en) 2013-02-08 2018-12-25 General Mills, Inc. Reduced sodium food products
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10702571B2 (en) 2015-12-03 2020-07-07 The University Of North Carolina At Pembroke Materials for cathepsin B enhancement and methods of use
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US11065224B2 (en) 2016-07-22 2021-07-20 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
WO2021151169A1 (en) * 2020-01-31 2021-08-05 AusCann Group Holdings Ltd Cannabinoid composition and manufacturing method
WO2021151168A1 (en) * 2020-01-31 2021-08-05 AusCann Group Holdings Ltd Method of cannabinoid therapy
US11400065B2 (en) 2019-03-01 2022-08-02 Flamel Ireland Limited Gamma-hydroxybutyrate compositions having improved pharmacokinetics in the fed state
US11504347B1 (en) 2016-07-22 2022-11-22 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
WO2022256720A3 (en) * 2021-06-03 2023-01-19 Arcadia Medicine, Inc. Enantiomeric entactogen compositions and methods of their use
US11583510B1 (en) 2022-02-07 2023-02-21 Flamel Ireland Limited Methods of administering gamma hydroxybutyrate formulations after a high-fat meal
US11602512B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11602513B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11779557B1 (en) 2022-02-07 2023-10-10 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11845736B2 (en) 2021-10-01 2023-12-19 Empathbio, Inc. Prodrugs of MDMA, MDA, and derivatives thereof
US11896573B2 (en) 2020-07-24 2024-02-13 XWPharma Ltd. Pharmaceutical compositions and pharmacokinetics of a gamma-hydroxybutyric acid derivative
US11912680B2 (en) 2021-12-28 2024-02-27 Empathbio, Inc. Nitric oxide releasing prodrugs of MDA and MDMA
US11925710B2 (en) 2020-10-05 2024-03-12 XWPharma Ltd. Modified release compositions of a GAMMA-hydroxybutyric acid derivative
US11944597B2 (en) 2021-03-19 2024-04-02 XWPharma Ltd. Pharmacokinetics of combined release formulations of a gamma-hydroxybutyric acid derivative

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6228863B1 (en) * 1997-12-22 2001-05-08 Euro-Celtique S.A. Method of preventing abuse of opioid dosage forms
US6309668B1 (en) * 1994-02-01 2001-10-30 Aventis Pharma Limited Abuse resistant tablets

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6309668B1 (en) * 1994-02-01 2001-10-30 Aventis Pharma Limited Abuse resistant tablets
US6228863B1 (en) * 1997-12-22 2001-05-08 Euro-Celtique S.A. Method of preventing abuse of opioid dosage forms

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8329744B2 (en) 2005-11-02 2012-12-11 Relmada Therapeutics, Inc. Methods of preventing the serotonin syndrome and compositions for use thereof
US9125833B2 (en) 2005-11-02 2015-09-08 Relmada Therapeutics, Inc. Multimodal abuse resistant and extended release opioid formulations
US8920838B2 (en) 2006-08-03 2014-12-30 Horizon Pharma Ag Delayed-release glucocorticoid treatment of rheumatoid disease
US9504699B2 (en) 2006-08-03 2016-11-29 Hznp Limited Delayed-release glucocorticoid treatment of rheumatoid disease
WO2009112874A1 (en) * 2008-03-10 2009-09-17 Eurodrug Laboratories B.V. Modified release composition comprising doxofylline
WO2009112436A1 (en) * 2008-03-10 2009-09-17 Eurodrug Laboratories B.V. Modified release composition comprising doxofylline
AU2009224801B2 (en) * 2008-03-10 2014-06-05 Pacific Healthcare (Thailand) Co. Ltd Modified release composition comprising doxofylline
US20120100212A1 (en) * 2009-07-06 2012-04-26 Kyorin Pharmaceutical, Co., Ltd. Tablet having hollow structure
AT509000B1 (en) * 2009-10-23 2012-12-15 Rausch Peter WATER-SOLUBLE PREPARATIONS OF CANNABINOIDS AND CANNABIC PREPARATIONS AND THEIR APPLICATIONS
US9125867B2 (en) 2010-02-24 2015-09-08 Invincible Biotechnology Diversion- and/or abuse-resistant compositions and methods for making the same
US20130098286A1 (en) * 2010-07-06 2013-04-25 Catherine Herry Method and agent for detecting drugs in beverages
AU2011278140B2 (en) * 2010-07-06 2015-07-09 Ethypharm Pharmaceutical form for combating chemical submission of a medicament
CN103118667A (en) * 2010-07-06 2013-05-22 埃迪制药公司 Pharmaceutical form for combating chemical submission of a medicament
CN103119435A (en) * 2010-07-06 2013-05-22 埃迪制药公司 Method and agent for detecting drugs in beverages
CN103118667B (en) * 2010-07-06 2015-09-16 埃迪制药公司 For hitting the medicament forms that pharmaceutical chemistry is presented
AU2011281482B2 (en) * 2010-07-06 2015-10-01 Ethypharm Method and agent for detecting drugs in beverages
US20130108556A1 (en) * 2010-07-06 2013-05-02 Ethypharm Pharmaceutical form for combating chemical submission of a medicament
KR101957657B1 (en) * 2010-07-06 2019-03-12 에씨팜 Method and agent for detecting drugs in beverages
KR20180080374A (en) * 2010-07-06 2018-07-11 에씨팜 Method and agent for detecting drugs in beverages
US11540539B2 (en) 2013-02-08 2023-01-03 General Mills, Inc. Reduced sodium food products
US10159268B2 (en) 2013-02-08 2018-12-25 General Mills, Inc. Reduced sodium food products
US10639281B2 (en) 2013-08-12 2020-05-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10792254B2 (en) 2013-12-17 2020-10-06 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US10702571B2 (en) 2015-12-03 2020-07-07 The University Of North Carolina At Pembroke Materials for cathepsin B enhancement and methods of use
US11826335B2 (en) 2016-07-22 2023-11-28 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11766418B2 (en) 2016-07-22 2023-09-26 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11504347B1 (en) 2016-07-22 2022-11-22 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11896572B2 (en) 2016-07-22 2024-02-13 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11839597B2 (en) 2016-07-22 2023-12-12 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11065224B2 (en) 2016-07-22 2021-07-20 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11602512B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11602513B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11400065B2 (en) 2019-03-01 2022-08-02 Flamel Ireland Limited Gamma-hydroxybutyrate compositions having improved pharmacokinetics in the fed state
WO2021151169A1 (en) * 2020-01-31 2021-08-05 AusCann Group Holdings Ltd Cannabinoid composition and manufacturing method
WO2021151168A1 (en) * 2020-01-31 2021-08-05 AusCann Group Holdings Ltd Method of cannabinoid therapy
US11896573B2 (en) 2020-07-24 2024-02-13 XWPharma Ltd. Pharmaceutical compositions and pharmacokinetics of a gamma-hydroxybutyric acid derivative
US11925710B2 (en) 2020-10-05 2024-03-12 XWPharma Ltd. Modified release compositions of a GAMMA-hydroxybutyric acid derivative
US11944597B2 (en) 2021-03-19 2024-04-02 XWPharma Ltd. Pharmacokinetics of combined release formulations of a gamma-hydroxybutyric acid derivative
WO2022256720A3 (en) * 2021-06-03 2023-01-19 Arcadia Medicine, Inc. Enantiomeric entactogen compositions and methods of their use
US11939312B2 (en) 2021-06-03 2024-03-26 Arcadia Medicine, Inc. Enantiomeric entactogen compositions and their use
US11845736B2 (en) 2021-10-01 2023-12-19 Empathbio, Inc. Prodrugs of MDMA, MDA, and derivatives thereof
US11912680B2 (en) 2021-12-28 2024-02-27 Empathbio, Inc. Nitric oxide releasing prodrugs of MDA and MDMA
US11779557B1 (en) 2022-02-07 2023-10-10 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11583510B1 (en) 2022-02-07 2023-02-21 Flamel Ireland Limited Methods of administering gamma hydroxybutyrate formulations after a high-fat meal

Also Published As

Publication number Publication date
WO2008033351A3 (en) 2009-04-09

Similar Documents

Publication Publication Date Title
WO2008033351A2 (en) Multimodal abuse resistant and extended release formulations
US9125833B2 (en) Multimodal abuse resistant and extended release opioid formulations
US8329744B2 (en) Methods of preventing the serotonin syndrome and compositions for use thereof
US20090082466A1 (en) Abuse Resistant and Extended Release Formulations and Method of Use Thereof
EP2448406B1 (en) Extended release oral pharmaceutical compositions of 3-hydroxy-n-methylmorphinan and method of use
WO2008024490A2 (en) Oral pharmaceutical formulations of abuse deterrent cannabinoids and method of use
US20110097395A1 (en) Oral Pharmaceutical Compositions of Buprenorphine and Method of Use
JP5730572B2 (en) Abuse resistant formulation
WO2008021394A2 (en) Pharmaceutical formulations of cannabinoids and method of use
WO2008027442A2 (en) Abuse deterrent oral pharmaceutical formulations of opioid agonists and method of use
JP6696994B2 (en) Immediate release abuse deterrent granule form
EP1878444A1 (en) Composition containing antidementia agent
US20100210732A1 (en) Methods of Preventing the Serotonin Syndrome and Compositions for Use Therefor
AU2005309406A1 (en) Methods and compositions for deterring abuse of orally administered pharmaceutical products
AU703242B2 (en) Film coated tablet of paracetamol and domperidone
WO2020188568A1 (en) Methods and compositions for treating autism spectrum disorder and associated disorders
JP2019070013A (en) Compositions and methods for reducing overdose
US20040006072A1 (en) Sustained-release alprazolam composition
WO2020183455A1 (en) Cannabinoid combinations for treating low back pain
EP2405754A1 (en) Modified release pharmaceutical compositions of buprenorphine
Reddy et al. Preparation and Characterization of Orodispersible tablets of Antispasmodic drug manufactured with Co-Processed mixtures
CA2847781C (en) Reducing drug liking in a subject
WO2020188569A1 (en) Methods and compositions for preventing or treating weight gain caused by psychiatric drugs
WO2020183457A1 (en) Cannabinoid combinations for treating diabetic neuropathy
EP3946267A1 (en) Pharmaceutical combination formulations comprising tizanidine, resveratrol and piperine

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07838038

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07838038

Country of ref document: EP

Kind code of ref document: A2