CA2690829A1 - Formulations of nonopioid and confined opioid analgesics - Google Patents

Formulations of nonopioid and confined opioid analgesics Download PDF

Info

Publication number
CA2690829A1
CA2690829A1 CA2690829A CA2690829A CA2690829A1 CA 2690829 A1 CA2690829 A1 CA 2690829A1 CA 2690829 A CA2690829 A CA 2690829A CA 2690829 A CA2690829 A CA 2690829A CA 2690829 A1 CA2690829 A1 CA 2690829A1
Authority
CA
Canada
Prior art keywords
hours
composition
acetaminophen
hydrocodone
released
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2690829A
Other languages
French (fr)
Inventor
Joerg Rosenberg
Gerd Woehrle
Thomas Y. Kessler
Joerg Breitenbach
Salih Durak
Friedrich W. Richter
Sandeep Dutta
Wei Liu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Deutschland GmbH and Co KG
Original Assignee
Abbott Gmbh & Co. Kg
Joerg Rosenberg
Gerd Woehrle
Thomas Y. Kessler
Joerg Breitenbach
Salih Durak
Friedrich W. Richter
Sandeep Dutta
Wei Liu
Abbvie Deutschland Gmbh & Co Kg
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 Abbott Gmbh & Co. Kg, Joerg Rosenberg, Gerd Woehrle, Thomas Y. Kessler, Joerg Breitenbach, Salih Durak, Friedrich W. Richter, Sandeep Dutta, Wei Liu, Abbvie Deutschland Gmbh & Co Kg filed Critical Abbott Gmbh & Co. Kg
Publication of CA2690829A1 publication Critical patent/CA2690829A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/2853Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers, poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • A61K9/2866Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse

Abstract

The preferred exemplary embodiments in the present application provide formulations and methods for the delivery of drugs, particularly drugs of abuse, having an abuse-relevant drug substantially confined in the core and a non-abuse relevant drug in a non-core region. These formulations have reduced potential for abuse. In the formulation, preferably the abuse relevant drug is an opioid and the non-abuse relevant drug is acetaminophen or ibuprofen.
More preferably, the opioid is hydrocodone, and the non-abuse relevant analgesic is acetaminophen. In certain preferred embodiments, the dosage forms are characterized by resistance to solvent extraction; tampering, crushing or grinding. Certain embodiments of the inventions provide dosage forms that provide an initial burst of release of drug followed by a prolonged period of controllable drug release.

Description

FORMULATIONS OF NONOPIOID AND CONFINED OPIOID ANALGESICS
TECHNICAL FIELD OF INVENTION
The present invention relates to compositions for oral administration.
Preferably the invention teaches at least one abuse-resistant composition for delivering a drug having an abuse potential, related methods of preparing these dosage forms, and methods of treating a patient in need thereof comprising administering the inventive compositions to the patient.
More preferably, these compositions include at least one non-opioid analgesic and at least one confined opioid analgesic.
BACKGROUND OF THE INVENTION
Abuse of prescription drugs has become a public health problem in many communities. Opioids are one common class of drugs that is subject to abuse. Opioids are the major class of analgesics used in the management of moderate to severe pain in the United States of America because of their effectiveness, ease of titration, and favorable risk-to-benefit ratio.
One of the effects of opioid administration is the ability of such drugs in some individuals to alter mood and feeling in a manner so as to provide a desirable sense of "well-being" dissociated from therapeutic ameliorative effects. Repeated illicit abuse further results in certain users being addicted to opioids. Similar to the opioids, many other classes of drugs are also subject to abuse, although the patterns and effects of the abuse vary.
Accordingly, in the art various methods and formulations have been described to diminish or eliminate various patterns of abuse, such as related to accidental or intentional dose dumping in alcohol, crushing and snorting, etc.
U.S. Patent Application 11/625,705 and PCT Application PCT/US07/60864 filed on January 22, 2007, which are incorporated herein by reference in their entirety for all purposes, describe various methods and compositions of abuse resistant formulations having drugs of abuse. In these patent applications, an extensive formulation screening program was used to identify suitable extrudate formulations exhibiting biphasic in vitro drug dissolution (> 30% after 1 h, >
80% after 8 h) for the narcotic drug hydrocodone bitartrate 2.5-hydrate. It was found, however, that the drug dissolution of the second agent did not meet the above criterion for biphasic drug dissolution (with > 30% after 1 h, > 80% after 8 h) with respect to acetaminophen, a.k.a.

paracetamol or APAP. Although both drugs, hydrocodone-bitartrate 2.5-hydrate and acetaminophen, were extruded and calendered from a homogeneously blended mixture of solids, all the studies on the resulting dosage forms showed that the two active ingredients were released at different rates. These in vitro data were also confirmed in experimental animal studies (minipig) and in a clinical study performed with these dosage forms. The clinical study also showed that although the desired kinetics were achieved for the hydrocodone bitartrate 2.5-hydrate, this was not the case for the acetaminophen. New formulation concepts therefore had to be found to achieve the required biphasic drug dissolution profile for the acetaminophen as well.
Further, it was also found that in most cases the calendered extrudate tablets manufactured in accordance with U.S.l 1/625,705 and PCT/US07/60864 patent applications had rough surfaces and therefore based of their appearance did not in all cases meet the criteria for marketable tablets. A need for improvement was thus also perceived in this respect.
While numerous compositions, formulations and methodologies exist to address abuse of drugs, all compositions, formulations and methods have limitations to a greater or lesser extent.
Accordingly, there is a need for providing new and/or improved formulations, compositions and methods of preventing abuse of drugs having abuse potential. More specifically, there is a need to develop oral formulations that would meet the biphasic drug dissolution profile and also have attributes that include drug deterrence and desirable appearance to meet the criteria for a marketable tablet.
This background information is provided for the purpose of making known some information believed by the applicant to be of possible relevance to the present invention. No admission is intended, nor should be construed, that any of the preceding information constitutes prior art to the present invention.

SUMMARY OF THE INVENTION
Certain preferred embodiments of the present invention provide dosage forms and methods for the delivery of drugs, particularly drugs of abuse, characterized by resistance to solvent extraction; tampering, crushing or grinding, and providing an initial burst of release of drug followed by a prolonged period of controllable drug release. Preferably, the dosage form includes at least one non-opioid analgesic and at least one confined opioid analgesic.
In one preferred embodiment, the present invention provides a pharmaceutical composition having a core and a non-core layer, comprising: (a) hydrocodone, a pharmaceutically acceptable salt or a hydrate thereof, and (b) acetaminophen or ibuprofen. In this embodiment, at least 75% all of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core, and the acetaminophen or the ibuprofen is the non-core layer. Further, this composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily. Preferably, greater than 90% of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core. More preferably, substantially all of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core. In another embodiment, the core further comprises acetaminophen or ibuprofen. More preferably, the core further comprises acetaminophen.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose. In another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose. In yet another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6 ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose. Other embodiments of the dosage form include about 5-20 mg of hydrocodone bitartrate pentahemihydrate and about 400-600 mg of acetaminophen. Yet another embodiment of the dosage form includes 10-15 mg of hydrocodone bitartrate pentahemihydrate and about 500-600 mg of acetaminophen.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. When administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg. In another embodiment, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg. In yet another embodiment, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
Preferably in this embodiment, the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone bitartrate pentahemihydrate.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. The dosage form produces a plasma concentration at 1 hour (Cl) for hydrocodone of about 0.18 ng/mL/mg to about 1.51 ng/mL/mg, and a plasma concentration at 1 hour C l for acetaminophen of about 2.34 ng/mL/mg to about 7.24 ng/mL/mg. In preferred embodiments such as Formulation 15, the dosage form produces a Cl for hydrocodone of about 0.32 ng/mL/mg to about 1.51 ng/mL/mg and a Cl for acetaminophen of about 2.34 ng/mL/mg to about 5.50 ng/mL/mg.
In certain other embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. The dosage form produces a plasma concentration at 1 hour (Cl) for hydrocodone from about 0.30 ng/mL/mg to about 1.06 ng/mL/mg, and a C l for acetaminophen from about 2.75 ng/mL/mg to about 5.57 ng/mL/mg. In preferred embodiments, the dosage from produces a Cl for hydrocodone from about 0.45 ng/mL/mg to about 1.06 ng/mL/mg and a C l for acetaminophen from about 2.75 ng/mL/mg to about 4.43 ng/mL/mg.
In other embodiments, the dosage form produces a combined C l for hydrocodone and acetaminophen from about 1.18 g/mL to about 3.63 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen, on fasting. In preferred embodiments, the dosage from produces a combined C l for hydrocodone and acetaminophen from about 1.18 g/mL to about 2.76 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.

In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.79 g/mL, after a single dose of 15 mg hydrocone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined C l for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.23 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In preferred embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen of 1.80 0.42 g/mL with the 95% confidence interval for the mean value falling between about 1.61 g/mL to about 2.00 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. The 95%
confidence interval of combined Cl for hydrocodone and acetaminophen for the preferred embodiments and the Control overlapped. The 95% confidence interval for the mean value of combined Cl for hydrocodone and acetaminophen for the Control ranged from about 1.46 to 1.96 g/mL, after administered as a single dose of 15 mg hydrocodone and 500 mg of acetaminophen to the human patient. The Control provides sufficient plasma levels of opioid and nonopioid analgesic to provide a reduction in pain intensity within about 1 hour after administration.
When administered to a population of healthy North Americans or Western Europeans, particularly when the formulation is adapted to be suitable for, or intended for, administration to a human every 12 hours as needed, about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C. In another embodiment, about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C.
Further, in another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60%
to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours. In another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and at least 90%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours. In another embodiment, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and at least 95%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours. Yet in another embodiment, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours. In another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 1 lhours to about 12 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours. In yet another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
However, when the a slow-release version of the formulation is adapted to be suitable for, or intended for administration to a human, twice daily, as needed, then at least 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours. In another embodiment of the slow release formulation, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours. In another embodiment of the slow release formulation, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours. In another embodiment of this slow release embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours. In yet another embodiment of the slow release formulation, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
In a preferred embodiment, the present invention provides a composition where the core layer comprises an excipient or a mixture of excipients capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug.
Further, in a preferred embodiment, the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer.
Most preferably, the composition comprises about 500mg of acetaminophen and about 15 mg of hydrocodone bitartrate pentahemihydrate.
In another exemplary embodiment, the present invention provides a pharmaceutical composition having a core and a non-core layer, comprising: (a) an abuse-relevant drug, a pharmaceutically acceptable salt or a hydrate thereof and a non-abuse-relevant drug or a pharmaceutically acceptable salt thereof in the core layer, and (b) a non-abuse-relevant drug, a pharmaceutically acceptable salt or a hydrate thereof in the non-core layer. Preferably, this composition is characterized by at least one of the following features:
i) the amount of abuse-relevant drug that is extracted from the composition by 40% aqueous ethanol within one hour at 37 C in vitro is less than or equal 1.5 times the amount of the abuse-relevant drug that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the abuse-relevant drug and not more than 45% of the abuse-relevant drug during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the non-abuse relevant drug within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the non-abuse relevant drug and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the abuse-relevant drug upon grinding increases by less than 2-to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37 C , vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0. 1 to about 0.2.
In this composition, the amount of the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 C is about 70% to about 130% of the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C. In another embodiment, the amount of the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 C is about 70% to about 90% of the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C.
In yet another embodiment, the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 C is about 75% to about 90% of the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C.
Another embodiment of the present invention provides a pharmaceutical composition having a core layer and a non-core layer. In this composition the core layer comprises a mixture of: (a) at least one opioid; and (b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof. The non-core layer comprises at least one non-opioid analgesic. Further, these compositions are adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily. Preferably, the core layer further comprises at least one non-opioid analgesic. In a preferred embodiment, the composition is characterized by at least one of the following features:
i) the amount of abuse-relevant drug that is extracted from the composition by 40% aqueous ethanol within one hour at 37 C in vitro is less than or equal 1.5 times the amount of the abuse-relevant drug that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the abuse-relevant drug and not more than 45% of the abuse-relevant drug during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the non-abuse relevant drug within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the non-abuse relevant drug and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the abuse-relevant drug upon grinding increases by less than 2-to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37 C , vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0.1 to about 0.2.
In one embodiment, the opioid is selected from the group consisting of alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levophenacylmorphan, levorphanol, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbulphine, narceine, nicomorphine, norpipanone, opium, oxycodone, oxymorphone, papvretum, pentazocine, phenadoxone, phenazocine, phenomorphan, phenoperidine, piminodine, propiram, propoxyphene, sufentanil, tilidine, and tramadol, and salts, hydrates and mixtures thereof.
Further, the non-opioid analgesic is selected from the group consisting of acetaminophen, aspirin, fentaynl, ibuprofen, indomethacin, ketorolac, naproxen, phenacetin, piroxicam, sufentanyl, sunlindac, interferon alpha, and salts, hydrates and mixtures thereof. Preferably, the opioid is hydrocodone and the non-opioid analgesic is acetaminophen or ibuprofen. More preferably, the opioid is hydrocodone and the non-opioid analgesic is acetaminophen.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose. In another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose. In yet another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6 ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. When administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg. In another embodiment, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg. In yet another embodiment, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
Preferably in this embodiment, the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone bitartrate pentahemihydrate.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone of about 0.18 ng/mL/mg to about 1.51 ng/mL/mg, and a plasma concentration at 1 hour Cl for acetaminophen of about 2.34 ng/mL/mg to about 7.24 ng/mL/mg.
In preferred embodiments such as Formulation 15, the dosage form produces a C
l for hydrocodone of about 0.32 ng/mL/mg to about 1.51 ng/mL/mg and a Cl for acetaminophen of about 2.34 ng/mL/mg to about 5.50 ng/mL/mg.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone from about 0.30 ng/mL/mg to about 1.06 ng/mL/mg, and a Cl for acetaminophen from about 2.75 ng/mL/mg to about 5.57 ng/mL/mg. In preferred embodiments, the dosage from produces a Cl for hydrocodone from about 0.45 ng/mL/mg to about 1.06 ng/mL/mg and a C l for acetaminophen from about 2.75 ng/mL/mg to about 4.43 ng/mL/mg.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 3.63 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 2.76 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.79 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.23 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In preferred embodiments, the dosage form produces a combined C l for hydrocodone and acetaminophen of 1.80 0.42 g/mL with the 95% confidence interval for the mean value falling between about 1.61 g/mL to about 2.00 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. The 95%
confidence interval of combined Cl for hydrocodone and acetaminophen for the preferred embodiments and the Control overlapped. The 95% confidence interval for the mean value of combined Cl for hydrocodone and acetaminophen for the Control ranged from about 1.46 to 1.96 g/mL, after administered as a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen to the human patient. The Control provides sufficient plasma levels of opioid and nonopioid analgesic to provide a reduction in pain intensity within about 1 hour after administration.
When administered to a population of healthy North Americans or Western Europeans, particularly when the formulation is adapted to be suitable for, or intended for, administration to a human every 12 hours as needed, about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C. In another embodiment, about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C.
Further, in another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60%
to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours. In another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and at least 90%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours. In another embodiment, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and at least 95%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours. Yet in another embodiment, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours. In another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 1 lhours to about 12 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours. In yet another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.

However, when the a slow-release version of the formulation is adapted to be suitable for, or intended for administration to a human, twice daily, as needed, then at least 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours. In another embodiment of the slow release formulation, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours. In another embodiment of the slow release formulation, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours. In another embodiment of this slow release embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours. In yet another embodiment of the slow release formulation, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
In a preferred embodiment, the present invention provides a composition where the core layer comprises an excipient capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug. Further, in a preferred embodiment, the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer. Most preferably, the composition comprises about 500mg of acetaminophen and about 15 mg of hydrocodone bitartrate pentahemihydrate.
In another embodiment, the present invention provides a pharmaceutical composition having a core layer and a non-core layer. In this composition, the core layer comprises a mixture of (a) at least one opioid and at least one first non-opioid analgesic; (b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof. The non-core layer comprises at least one second non-opioid analgesic. Further, the composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily. In this embodiment, preferably, the opioid comprises hydrocodone and the first and the second non-opioid analgesic comprises acetaminophen or ibuprofen. More preferably, the opioid comprises hydrocodone and the first and the second non-opioid analgesic comprises acetaminophen.
Further, in this embodiment, the non-core layer comprises: (a) acetaminophen; and (b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof.
Preferably, the polymer or copolymer is selected from the group consisting of: hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose; polymethacrylate, polyvinyl alcohol, polyethylene oxide, and combinations thereof. More preferably, the polymer or copolymer is selected from the group consisting of: hydroxypropyl methylcellulose, and polyvinyl alcohol, or combinations thereof. Yet more preferably, the polymer or copolymer is selected from the group consisting of: polyvinyl alcohol and polyethylene oxide graft copolymers.
Further, in this embodiment, the ratio of acetaminophen to the rate controlling polymer or copolymer or combination thereof is about 1:1 to about 10:1. More preferably, the ratio of acetaminophen to the rate controlling polymer or copolymer or combination thereof is about 3:1 to about 5:1. As provided in the present invention, in one preferred embodiment, the non-core layer has at least one of the following characteristics:
(a) substantially does not crack after 3 months at 40 C, 75% relative humidity in induction-sealed HDPE bottles;
(b) substantially dry (not sticky);
provides fast dissolution in 0.01N HC1 at 37 C to expose the core layer releases at least 80% of the acetaminophen in the non-core layer within 20 minutes of administration to a human patient; or (e) provides a white pigmentation to the formulation without additional pigments.

In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose. In another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose. In yet another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6 ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. When administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg. In another embodiment, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg. In yet another embodiment, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
Preferably in this embodiment, the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone bitartrate pentahemihydrate.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone of about 0.18 ng/mL/mg to about 1.51 ng/mL/mg, and a plasma concentration at 1 hour C l for acetaminophen of about 2.34 ng/mL/mg to about 7.24 ng/mL/mg.
In preferred embodiments such as Formulation 15, the dosage form produces a C
l for hydrocodone of about 0.32 ng/mL/mg to about 1.51 ng/mL/mg and a Cl for acetaminophen of about 2.34 ng/mL/mg to about 5.50 ng/mL/mg.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone from about 0.30 ng/mL/mg to about 1.06 ng/mL/mg, and a Cl for acetaminophen from about 2.75 ng/mL/mg to about 5.57 ng/mL/mg. In preferred embodiments, the dosage from produces a Cl for hydrocodone from about 0.45 ng/mL/mg to about 1.06 ng/mL/mg and a C l for acetaminophen from about 2.75 ng/mL/mg to about 4.43 ng/mL/mg.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 3.63 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 2.76 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.79 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.23 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In preferred embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen of 1.80 0.42 g/mL with the 95% confidence interval for the mean value falling between about 1.61 g/mL to about 2.00 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. The 95%
confidence interval of combined Cl for hydrocodone and acetaminophen for the preferred embodiments and the Control overlapped. The 95% confidence interval for the mean value of combined Cl for hydrocodone and acetaminophen for the Control ranged from about 1.46 to 1.96 g/mL, after administered as a single dose of 15 mg hydrocodone and 500 mg of acetaminophen to the human patient. The Control provides sufficient plasma levels of opioid and nonopioid analgesic to provide a reduction in pain intensity within about 1 hour after administration.
When administered to a population of healthy North Americans or Western Europeans, particularly when the formulation is adapted to be suitable for, or intended for, administration to a human every 12 hours as needed, about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C. In another embodiment, about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C.
Further, in another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60%
to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours. In another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and at least 90%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours. In another embodiment, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and at least 95%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours. Yet in another embodiment, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours. In another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 1 lhours to about 12 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours. In yet another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
However, when the a slow-release version of the formulation is adapted to be suitable for, or intended for administration to a human, twice daily, as needed, then at least 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours. In another embodiment of the slow release formulation, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours. In another embodiment of the slow release formulation, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours. In another embodiment of this slow release embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours. In yet another embodiment of the slow release formulation, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
In a preferred embodiment, the present invention provides a composition where the core layer comprises an excipient capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug. Further, in a preferred embodiment, the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer. Most preferably, the composition comprises about 500mg of acetaminophen and about 15 mg of hydrocodone bitartrate pentahemihydrate.
These and other objects, advantages, and features of the invention will become apparent to those persons skilled in the art upon reading the details of the methods of the invention and compositions used therein as more fully described below.
BRIEF DESCRIPTION OF FIGURES:
Figure 1 depicts that coating the extrudated tablets resulted in significant smoothing of the tablet surface.
Figure 2 depicts schematics for calculation of Surface Roughness using Centre Line Average (CLA) approach.
Figure 3 depicts Centre Line Average (CLA) for an uncoated formulation. For uncoated formulation CLA = 36.1, when (N = 69).
Figure 4 depicts Centre Line Average (CLA) for an uncoated formulation. For a coated formulation CLA = 10.4, when (N = 69).

Figure 5 depicts preliminary mean hydrocodone concentration-time profiles for Formulations 15, and 16 and Control 1 for (a) 48 hours and (b) 12 hours.
Figure 6 depicts preliminary mean acetaminphen concentration-time profiles for Formulations 15, and 16 and Control 1 for (a) 48 hours and (b) 12 hours.
Figure 7 depicts in vitro drug release profiles for hydrocodone and acetaminphen for Formulations 17, and 18, Control 2 and uncoated Formulation VM-1 for 480 minutes.
DETAILED DESCRIPTION OF THE INVENTION
The invention is not limited to the particular methodology, protocols, animal studies, and reagents described, which can vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which will be limited only by the appended claims.
It must be noted that as used herein and in the appended claims, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise.
Thus, for example, reference to "a compound" includes a plurality of such compounds and equivalents thereof known to those skilled in the art, and so forth. As well, the terms "a" (or "an"), "one or more" and "at least one" can be used interchangeably herein. It is also to be noted that the terms "comprising", "including", and "having" can be used interchangeably.
Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs.
Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference for the purpose of describing and disclosing the chemicals, animals, instruments, statistical analysis and methodologies which are reported in the publications which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
Trademarks are used in this description as a convenient abbreviation for well known materials.
As one of ordinary skill would appreciate, the following brand names indicate the substances indicated:
EUDRAGIT : Polymers derived from esters of acrylic and methacrylic acid;

METHOCEL : Methyl or methoxyl Cellulose KOLLICOAT IR : Polyvinyl alcohol-polyethylene glycol-graft copolymers PLASDONE : Polyvinylpyrrolidone polymer or -copolymer LAUROGLYCOL : Propylene glycol laurate ester SPAN : Sorbitan fatty acid esters CREMOPHOR : Polyethoxylated Castor oil POLOXAMER : Polyoxyethylene polyoxypropylene block copolymers or polyoxyethylene polypropyleneglycol TWEEN : Polyethoxylated Sorbitan esters KLUCEL : Hydroxypropylcellulose KOLLIDON : Polyvinlypyrrolidone homo- or copolymers XYLITOL : (2,3,4,5)tetrahydroxy-pentanol ISOMALT : An equimolar composition of 6-0-a-D-glucopyranosido-D-sorbitol (1,6-GPS) and 1-0-a-D-glucopyranosido-D-mannitol-dihydrate (l,l-GPM-dihydrate).
POLYOX : Water-Soluble Resins based on polyethyleneoxide XYLIT : (2,3,4,5)tetrahydroxy-pentanol PLUROL OLEIQUE : Oleic esters of polyglycerol LUTROL : Polyoxyethylene polyoxypropylene block copolymers or polyoxyethylene polypropyleneglycol ETHOCEL : Ethylcellulose PRIMOJEL : Sodium starch glycolate The present invention provides an improved solid or solid solution, oral dosage formulation that provides for the in vivo sustained-release of pharmaceutically active compounds ("drugs") that have properties that make them likely to be abused or have been shown to be frequently abused, as well as salts, esters, prodrugs and other pharmaceutically-acceptable equivalents thereof.
The term "AUC" refers to the area under the concentration time curve, calculated using the trapezoidal rule and Clast/k, where Clast is the last observed concentration and k is the calculated elimination rate constant.
The term "AUCt" refers to the area under the concentration time curve to last observed concentration calculated using the trapezoidal rule.

The term "Cmax" refers to the plasma concentration of the referent abuse relevant drug at Tmax, expressed as ng/mL and g/mL, respectively, produced by the oral ingestion of a composition of the invention. Unless specifically indicated, Cmax refers to the overall maximum observed concentration.
The term "Cmin" refers to the minimum observed concentration within the intended dosing interval, e.g., a twelve hour dosing interval for a formulation labelled as suitable for dosing every 12 hours or as needed, of a dosage form of the invention administered for 5 doses contiguous dosing intervals.
The term "ng*hr/mL/mg" refers to the amount of the substance measured in nanograms times the number of hours per milliliter of blood divided by the milligrams of the abuse relevant drug administered to the animal or human.
As used herein, the phrase "ascending release rate" refers to a dissolution rate that generally increases over time, such that the drug dissolves in the fluid at the environment of use at a rate that generally increases with time, rather than remaining constant or decreasing, until the dosage form is depleted of about 80% of the drug.
When used in the above or other treatments, a therapeutically effective dose of one of the compounds of the present invention can be employed in pure form or, where such forms exist, in pharmaceutically acceptable salt, ester or prodrug form. The phrase "therapeutically effective dose" of the compound includes of the invention means a sufficient amount of the compound to treat disorders, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder;
activity of the specific compound employed; the specific composition employed;
the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
In one preferred embodiment, the invention provides dosage forms that inhibit the extraction of the drug by common solvents, e.g., without limitation, distilled aqueous ethanol, from the formulation. The formulation dissuades abuse by limiting the ability of persons to extract the opioid from the formulation (either intentionally or unintentionally), such that the opioid cannot easily be concentrated for parenteral administration. Also these abuse resistant formulations may not be easily broken down into smaller particulates or powder-form that are easily abused by nasal snorting. Such an abuse-resistant formulation does not require incorporation of an opioid antagonist (albeit, an opioid antagonist may be added to the preparation to further dissuade abuse). While not desiring to be bound by any particular theory, it is believed that incorporation of alkylcelluloses, such as (without limitation) hydroxymethylcelluloses, and preferably hydroxypropylmethylcelluloses contribute to the formulation's resistance to extraction in alcohol, particularly in 20% or 40% aqueous ethanol. The alkylcellulose preferably has at least 12% substitution with an alkylsubstituent, more preferably at least 16%
substitution with an alkyl substituent, and most preferably at least 19% substitution with an alkyl substituent. Alkyl substitutions of the cellulose below about 40%, and more preferably below about 30%, are preferred in the context of the invention. Additionally, the alkyl substituent is preferably Ci-C6, more preferably Ci, Cz or C4, and most preferably C3, and can be straight-chained or branched when the alkyl substituent contains 3 or more carbon atoms.
In another preferred embodiment, the dosage forms optionally resists cutting, grinding, pulverization and the like. A convenient measure for this aspect of the invention is "breaking strength," as measured by "Pharma Test PTB 501" hardness tester. The inventive formulation preferably has a breaking strength of at least 150 newtons (150 N). More preferably, the inventive formulation has breaking strength of at least 300 N, yet more preferably of at least 450 N, and yet more preferably of at least 500 N.
Breaking strength according to the present invention can be determined with a tablet 10 mm in diameter and 5 mm in width according to the method for determining the breaking strength of tablets published in the European Pharmacopoeia 1997, page 143, 144, method no. 2.9.8. A
preferred apparatus used to measure breaking strength is a "Zwick Z 2.5"
materials tester, Fmax = 2.5 kN, draw max. 1150 mm with the set up comprising a column and a spindle, clearance behind of 100 mm, and a test speed of 0.1800 mm/min. Measurement can be performed using a pressure piston with screw-in inserts and a cylinder (10 mm diameter), a force transducer, (Fmax. 1 kN, diameter = 8 mm, class 0.5 from 10 N, class 1 from 2 N to ISO
7500-1, Zwick gross force Fmax = 1.45 kN). The apparatus can optionally be obtained from Zwick GmbH &
Co. KG, Ulm, Germany.
Any suitable means can be used to produce the inventive composition. In a preferred embodiment, the formulation is preferably melt-processed, and more preferably melt-extruded, and then in either case directly shaped without milling or grinding the formulation.
Notwithstanding the foregoing, it is contemplated that the directly shaped tablets of the formulation can be optionally coated with a swallowing aid, such as without limitation, a gelatin coat. While not desiring to be bound by any particular theory, it is believed that direct shaping to prevent undesirable sharp features from forming on the formulation without an intermediate grinding step contributes to the superior breaking strength of the formulation. Additionally, embodiments of the inventive formulation optionally gain additional breaking strength by employing at least two melt-processed polymers. While not ascribing to any particular theory, it is believed that the second melt-processed polymer preferentially interacts with the first melt-processed polymer so as to advantageously adjust the transition glass temperature of the composition as a whole during the formation of the tablet.
In one embodiment, the formulation may use a polymer, or a copolymer, or a combination thereof to create the melt-processed, and more preferably melt-extruded, directly shaped formulation. Polymers that are pharmacologically inactive and provide enteric coatings or sustained release profile for the formulation can also be used. In one embodiment, suitable polymers/copolymers include poly(meth)acrylate like e.g. Eudragit L- or S-type, which are pharmacologically inactive.
EUDRAGIT is a tradename for some preferred polymers that are suitable for use in the invention and are derived from esters of acrylic and methacrylic acid. The properties of the EUDRAGIT polymers are principally determined by functional groups incorporated into the monomers of the EUDRAGIT polymers. The individual EUDRAGIT grades differ in their proportion of neutral, alkaline or acid groups and thus in terms of physicochemical properties.
Ammonioalklyl methacrylate copolymers or methacrylate copolymers may be used having the following formula:

iH3 (H) iH3 C~ C
Alkyl-OOC R

The Eudragit polymers fulfil the specifications/requirements set in the USP.
According to 2007 US Pharmacopoeia, Eudragit is defined as USP 30 / NF 25.
Methacrylic acid copolymer, type A NF = Eudragit L- 100 Methacrylic acid copolymer, type B NF = Eudragit S-100 Methacrylic acid copolymer, type C NF = Eudragit L-100-55 (contains a small detergent amount) Ammonio Methacrylate Copolymer, type A NF = Eudragit RL- 100 (granules) Ammonio Methacrylate Copolymer, type A NF = Eudragit RL-PO (powder) Ammonio Methacrylate Copolymer, type B NF = Eudragit RS-100 (granules) Ammonio Methacrylate Copolymer, type B NF = Eudragit RS-PO (powder) Polyacrylate Dispersion 30 Percent Ph. Eur. = Eudragit NE30D (= 30% aqueous dispersion) Basic butylated methacrylate copolymer Ph. Eur. = Eudragit E- 100 wherein the functional group has a quatemary ammonium (trimethylammonioethyl methacrylate) moiety or R = COOCH2CH2N+(CH3)3C1- [commercially available as EUDRAGIT (RL or RS)]
or the functional group is a carboxylic acid, or R = COOH [commercially available as EUDRAGIT (L)]. When the functional group is a carboxylic acid moiety, the EUDRAGIT
(L) polymer is gastroresistant and enterosoluble. Thus formulations using EUDRAGIT (L) will be resistant to gastric fluid and will release the active agent in the colon. When the functional group is a trimethylammonioethyl methacrylate moiety, the EUDRAGIT
(RL or RS) polymers are insoluble, permeable, dispersible and pH-independent. These EUDRAGIT (RL
or RS) polymers may therefore be used for delayed drug release for sustained release formulations. EUDRAGIT is sold in various forms such as in solid form (EUDRAGIT
L100/ S100/ L-100-55, EUDRAGIT E PO, EUDRAGIT RL PO, Eudragit RS PO), granules (EUDRAGIT E100, EUDRAGIT RL 100/RS 100), dispersions (L 30 D-55/FS 30D 30%, EUDRAGIT NE 30 D/40 D 30%/40% polymer content, EUDRAGIT RL 30 D RS 30 D 30%) and organic solutions (EUDRAGIT L 12.5, EUDRAGIT E12.5, EUDRAGIT RL 12.5/RS
12.5 - 12.5% organic solution).
When at least two melt-processed polymers are employed, one is preferably a cellulose derivative, more preferably a hydroxyalkylcellulose derivative, and optionally hydroxypropylmethylcellulose, and independently, the other polymer is preferably a (meth)acrylate polymer (such as, any suitable Eudragit polymer). Among the (meth)acrylate polymer polymers preferred in the context of the invention are Eudragit L and Eudragit RS. One more preferred polymer in the context of the invention is Eudragit RL. The Eudragit polymers can be used in combinations, with mixtures of Eudragit RS and RL being preferred.
Persons that (albeit inadvisedly) drink substantial quantities of alcoholic beverages when taking physician prescribed medications can substantially alter the composition of the gastric juices contained in the stomach, and in extreme cases these gastric juices can comprise up to 40%
alcohol. Advantageously, embodiments of the inventive abuse-deterrent formulation optionally comprises a melt-processed mixture of at least one abuse-relevant drug, at least one cellulose ether or cellulose ester, and at least one (meth)acrylic polymer, wherein the amount of the drug that is extracted from the formulation by 20% aqueous ethanol, or 40% aqueous ethanol, or both, within one hour at 37 C is less than or equal 1.5 times the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C, or at 25 C or both. The resistance to extraction by 40% ethanol is advantageous in those situations in which an individual purposefully attempts to extract an abuse relevant drug from a medicine containing an abuse relevant drug.
The protocols for extraction by 20% or 40% aqueous ethanol or 0.01 N
hydrochloric acid, respectively, are given in the experimental section that follows. In more preferred embodiments, the amount of the drug that is extracted from the formulation by 20% or 40%
aqueous ethanol is less than or equal 1.5 times the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour. In a yet more preferred embodiments, the amount of the drug that is extracted from the formulation by 20% or 40% aqueous ethanol is less than or equal the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour. In a yet more preferred embodiments, the amount of the drug that is extracted from the formulation by 20% or 40%
aqueous ethanol is less than or equa10.9 times the amount of the drug that is extracted by 0.01 N
hydrochloric acid within one hour.

The present invention also provides a sustained release formulation of at least one abuse relevant drug that hampers the extraction of the drug from the formulation when extraction is by solvent extraction with commonly available household extraction solvents such as isopropyl alcohol, distilled alcohols exemplified by vodka, white vinegar, water and aqueous ethanol (e.g., 20%
ethanol). Whereas the formulation is largely resistant to solvent-extraction, it still provides adequate drug release in aqueous solutions such as gastric fluids. This formulation when crushed or ground also provides adequate drug release in aqueous solutions such as gastric fluids.
Fortunately, in certain preferred embodiments of the invention, the amount of the abuse relevant drug released from the time of placing in 3 oz. of one, or two, or three, or more than three, of the household solvents listed above (i.e., 0 hours) to 1 hour is expected to be not more than 15%
greater than the amount released over the same time as when swallowed by an ordinary human, or the more than 1 hour to about 4 hours is not more than 15% greater than the amount released over the same time as when swallowed by an ordinary human, or both.
Exemplary preferred compositions of the invention comprise cellulose ethers and cellulose esters, which can be used alone or in combination in the invention have a preferable molecular weight in the range of 50,000 to 1,250,000 daltons. Cellulose ethers are preferably selected from alkylcelluloses, hydroxalkylcelluloses, hydroxyalkyl alkylcelluloses or mixtures therefrom, such as ethylcellulose, methylcellulose, hydroxypropyl cellulose (NF), hydroxyethyl cellulose (NF), and hydroxpropyl methylcellulose (USP), or combinations thereof. Useful cellulose esters are, without limitation, cellulose acetate (NF), cellulose acetate butyrate, cellulose acetate propionate, hydroxypropylmethyl cellulose phthalate, hydroxypropylmethyl cellulose acetate phthalate, and mixtures thereof. Most preferably, non-ionic polymers, such as hydroxypropylmethyl cellulose may be used.
The amount of substituent groups on the anhydroglucose units of cellulose can be designated by the average number of substituent groups attached to the ring, a concept known to cellulose chemists as "degree of substitution" (D. S.). If all three available positions on each unit are substituted, the D. S. is designated as 3, if an average of two on each ring are reacted, the D. S. is designated as 2, etc.
In preferred embodiments, the cellulose ether has an alkyl degree of substitution of 1.3 to 2.0 and hydroxyalkyl molar substitution of up to 0.85.

In preferred embodiments, the alkyl substitution is methyl. Further, the preferred hydroxyalkyl substitution is hydroxpropyl. These types of polymers with different substitution degrees of methoxy- and hydroxypropoxy-substitutions are summarized listed in pharmacopoeas, e.g. USP
under the name "Hypromellose".

Methylcellulose is available under the brand name METHOCEL A. METHOCEL A has a methyl (or methoxyl) D. S. of 1.64 to 1.92. These types of polymers are listed in pharmacopoeas, e.g. USP under the name "Methylcellulose".
A particularly preferred cellulose ether is hydroxpropyl methylcellulose.
Hydroxpropyl methylcellulose is available under the brand name METHOCEL E (methyl D. S.
about 1.9, hydroxypropyl molar substitution about 0.23), METHOCEL F (methyl D. S. about 1.8, hydroxypropyl molar substitution about 0.13), and METHOCEL K (methyl D. S.
about 1.4, hydroxypropyl molar substitution about 0.21). METHOCEL F and METHOCEL K are preferred hydroxpropyl methylcelluloses for use in the present invention.
The acrylic polymer suitably includes homopolymers and copolymers (which term includes polymers having more than two different repeat units) comprising monomers of acrylic acid and/or alkacrylic acid and/or an alkyl (alk)acrylate. As used herein, the term "alkyl (alk)acrylate"
refers to either the corresponding acrylate or alkacrylate ester, which are usually formed from the corresponding acrylic or alkacrylic acids, respectively. In other words, the term "alkyl (alk)acrylate" refers to either an alkyl alkacrylate or an alkyl acrylate.
Preferably, the alkyl (alk)acrylate is a(Ci-Czz)alkyl ((Ci-Cio)alk)acrylate.
Examples of Ci-Czz alkyl groups of the alkyl (alk)acrylates include methyl, ethyl, n-propyl, n-butyl, iso-butyl, tert-butyl, iso-propyl, pentyl, hexyl, cyclohexyl, 2-ethyl hexyl, heptyl, octyl, nonyl, decyl, isodecyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl, octadecyl, nonadecyl, eicosyl, behenyl, and isomers thereof. The alkyl group may be straight or branched chain.
Preferably, the (Ci-Czz)alkyl group represents a(Ci-C6)alkyl group as defined above, more preferably a(Ci-C4)alkyl group as defined above. Examples of Ci-io alk groups of the alkyl (alk)acrylate include methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, pentyl, hexyl, cyclohexyl, 2-ethyl hexyl, heptyl, octyl, nonyl, decyl and isomers thereof. The alk groups may be straight or branched chain. Preferably, the (Ci-Cio)alk group represents a(Ci-C6)alk group as defined above, more preferably a(Ci-C4) alk group as defined above.

Preferably, the alkyl (alk)acrylate is a(Ci-C4)alkyl ((Ci-C4) alk)acrylate, most preferably a(Ci-C4)alkyl (meth)acrylate. It will be appreciated that the term (Ci-C4)alkyl (meth)acrylate refers to either (Ci-C4)alkyl acrylate or (Ci-C4)alkyl methacrylate. Examples of (Ci-C4)alkyl (meth)acrylate include methyl methacrylate (MMA), ethyl methacrylate (EMA), n-propyl methacrylate (PMA), isopropyl methacrylate (IPMA), n-butyl methacrylate (BMA), isobutyl methacrylate (IBMA), tert-butyl methacrylate (TBMA): methyl acrylate (MA), ethyl acrylate (EA), n-propyl acrylate (PA), n-butyl acrylate (BA), isopropyl acrylate (IPA), isobutyl acrylate (IBA), and combinations thereof.
Preferably, the alkacrylic acid monomer is a(Ci-Cio)alkacrylic acid. Examples of (Ci-Cio)alkacrylic acids include methacrylic acid, ethacrylic acid, n-propacrylic acid, iso-propacrylic acid, n-butacrylic acid, iso-butacrylic acid, tert-butacrylic acid, pentacrylic acid, hexacrylic acid, heptacrylic acid and isomers thereof. Preferably the (Ci-Cio)alkacrylic acid is a(Ci-C4)alkacrylic acid, most preferably methacrylic acid.
In certain embodiments, the alkyl groups may be substituted by aryl groups. As used herein "alkyl" group refers to a straight chain, branched or cyclic, saturated or unsaturated aliphatic hydrocarbons. The alkyl group has 1-16 carbons, and may be unsubstituted or substituted by one or more groups selected from halogen, hydroxy, alkoxy carbonyl, amido, alkylamido, dialkylamido, nitro, amino, alkylamino, dialkylamino, carboxyl, thio and thioalkyl. A "hydroxy"
group refers to an OH group. An "alkoxy" group refers to an --O-alkyl group wherein alkyl is as defined above. A "thio" group refers to an --SH group. A "thioalkyl" group refers to an --SR
group wherein R is alkyl as defined above. An "amino" group refers to an --NH2 group. An "alkylamino" group refers to an --NHR group wherein R is alkyl is as defined above. A
"dialkylamino" group refers to an --NRR' group wherein R and R' are all as defined above. An "amido" group refers to an --CONH2. An "alkylamido" group refers to an --CONHR
group wherein R is alkyl is as defined above. A "dialkylamido" group refers to an --CONRR' group wherein R and R' are alkyl as defined above. A "nitro" group refers to an NOz group. A
"carboxyl" group refers to a COOH group.
In certain embodiments, the alkyl groups may be substituted by aryl groups. As used herein, "aryl" includes both carbocyclic and heterocyclic aromatic rings, both monocyclic and fused polycyclic, where the aromatic rings can be 5- or 6-membered rings.
Representative monocyclic aryl groups include, but are not limited to, phenyl, furanyl, pyrrolyl, thienyl, pyridinyl, pyrimidinyl, oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl and the like.
Fused polycyclic aryl groups are those aromatic groups that include a 5- or 6-membered aromatic or heteroaromatic ring as one or more rings in a fused ring system.
Representative fused polycyclic aryl groups include naphthalene, anthracene, indolizine, indole, isoindole, benzofuran, benzothiophene, indazole, benzimidazole, benzthiazole, purine, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,8-naphthyridine, pteridine, carbazole, acridine, phenazine, phenothiazine, phenoxazine, and azulene. Also as used herein, aryl group also includes an arylalkyl group. Further, as used herein "arylalkyl" refers to moieties, such as benzyl, wherein an aromatic is linked to an alkyl group.
Preferably, the acrylic polymer is an acrylic copolymer. Preferably, the acrylic copolymer comprises monomers derived from alkyl (alk)acrylate, and/or acrylic acid and/or alkacrylic acid as defined hereinbefore. Most preferably, the acrylic copolymer comprises monomers derived from alkyl (alk)acrylate, i.e. copolymerisable alkyl acrylate and alkyl alkacrylate monomers as defined hereinbefore. Especially preferred acrylic copolymers include a(Ci-C4)alkyl acrylate monomer and a copolymerisable (Ci-C4)alkyl (Ci-C4)alkacrylate comonomer, particularly copolymers formed from methyl methacrylate and a copolymerisable comonomer of methyl acrylate and/or ethyl acrylate and/or n-butyl acrylate.
Preferably, the (meth)acrylic polymer is a ionic (meth)acrylic polymer, in particular a cationic (meth)acrylic polymer. Ionic (meth)acrylic polymer are manufactured by copolymerising (meth)acrylic monomers carrying ionic groups with neutral (meth)acrylic monomers. The ionic groups preferably are quatemary ammonium groups.
The (meth)acrylic polymers are generally water-insoluble, but are swellable and permeable in aqueous solutions and digestive fluids. The molar ratio of cationic groups to the neutral (meth)acrylic esters allows for are control of the water-permeabilty of the formulation. In preferred embodiments the (meth)acrylic polymer is a copolymer or mixture of copolymers wherein the molar ratio of cationic groups to the neutral (meth)acrylic esters is in the range of about 1:20 to 1:35 on average. The ratio can by adjusted by selecting an appropriate commercially available cationic (meth)acrylic polymer or by blending a cationic (meth)acrylic polymer with a suitable amount of a neutral (meth)acrylic polymer.
Suitable (meth)acrylic polymers are commercially available from Rohm Pharma under the Tradename Eudragit, preferably Eudragit RL and Eudragit RS. Eudragit RL and Eudragit RS are copolymers of acrylic and methacrylic esters with a low content of quaternary ammonium groups, the molar ratio of ammonium groups to the remaining neutral (meth)acrylic esters being 1:20 in Eudragit RL and 1:40 in Eudragit RS. The mean molecular weight is about 150,000.
Besides the (meth)acrylic polymers, further pharmaceutically acceptable polymers may be incorporated in the inventive formulations in order to adjust the properties of the formulation and/or improve the ease of manufacture thereof. These polymers may be selected from the group comprising: homopolymers of N-vinyl lactams, especially polyvinylpyrrolidone (PVP), copolymers of a N-vinyl lactam and and one or more comonomers copolymerizable therewith, the comonomers being selected from nitrogen-containing monomers and oxygen-containing monomers; especially a copolymer of N-vinyl pyrrolidone and a vinyl carboxylate, preferred examples being a copolymer of N-vinyl pyrrolidone and vinyl acetate or a copolymer of N-vinyl pyrrolidone and vinyl propionate; polyvinyl alcohol-polyethylene glycol-graft copolymers (available as, e.g., Kollicoat IR from BASF AG, Ludwigshafen, Germany); high molecular polyalkylene oxides such as polyethylene oxide and polypropylene oxide and copolymers of ethylene oxide and propylene oxide; polyacrylamides; vinyl acetate polymers such as copolymers of vinyl acetate and crotonic acid, partially hydrolyzed polyvinyl acetate (also referred to as partially saponified "polyvinyl alcohol"); polyvinyl alcohol;
poly(hydroxy acids) such as poly(lactic acid), poly(glycolic acid), poly(3-hydroxybutyrate) and poly(3-hydroxybutyrate-co-3-hydroxyvalerate); or mixtures of one or more thereof. PVP
generates hydrocodone N-oxide during extrusion, therefore use of PVP-polymers and -copolymers is not always preferred. However, when a small amount (0.2 - 0.6 % w/w of the total formulation) of antioxidant is used, then PVP may be used preferably.
"Abuse-relevant drug" is intended to mean any biologically effective ingredient the distribution of which is subject to regulatory restrictions. Drugs of abuse that can be usefully formulated in the context of the invention include without limitation pseudoephedrine, anti-depressants, strong stimulants, diet drugs, steroids, and non-steroidal anti-inflammatory agents.
In the category of strong stimulants, methamphetamine is one drug that has recently received popular attention as a drug of abuse. There is also some concern at the present time about the abuse potential of atropine, hyoscyamine, phenobarbital, scopolamine, and the like. Another major class of abuse-relevant drugs are analgesics, especially the opioids.

By the term "opioid," it is meant a substance, whether agonist, antagonist, or mixed agonist-antagonist, which reacts with one or more receptor sites bound by endogenous opioid peptides such as the enkephalins, endorphins and the dynorphins. Opioids include, without limitation, alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levophenacylmorphan, levorphanol, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbulphine, narceine, nicomorphine, norpipanone, opium, oxycodone, oxymorphone, papvretum, pentazocine, phenadoxone, phenazocine, phenomorphan, phenoperidine, piminodine, propiram, propoxyphene, sufentanil, tilidine, and tramadol, and salts and mixtures thereof.
In some preferred embodiments, the inventive formulation includes at least one additional therapeutic drug. In even more preferred embodiments, the additional therapeutic dug can be, without limitation, selected from the group consisting of non-steroidal, non-opioidal analgesics, and is optionally further selected from the group consisting of acetaminophen, aspirin, fentaynl, ibuprofen, indomethacin, ketorolac, naproxen, phenacetin, piroxicam, sufentanyl, sunlindac, and interferon alpha. Particularly preferred are those combinations of drug currently sold as fixed dose combinations to the public under the authority of a suitable national or regional regulatory agency, such as (by way of example) the U.S. Food and Drug Administration.
Such drugs include without limitation a (fixed dose) combination of hydrocodone and acetaminophen, or a (fixed dose) combination of hydrocodone and ibuprofen.
The abuse-relevant drug(s) are preferably dispersed evenly throughout a matrix that is preferably formed by a cellulose ether or cellulose ester, and one acrylic or methacrylic polymer as well as other optional ingredients of the formulation. This description is intended to also encompass systems having small particles, typically of less than 1 m in diameter, of drug in the matrix phase. These systems preferably do not contain significant amounts of active opioid ingredients in their crystalline or microcrystalline state, as evidenced by thermal analysis (DSC) or X-ray diffraction analysis (WAXS). At least 98% (by weight) of the total amount of drug is preferably present in an amorphous state. If additional non-abuse relevant drug actives like e.g.
acetaminophen are additionally present in a formulation according to the present invention, this additional drug active(s) may be in a crystalline state embedded in the formulation.
When the dispersion of the components is such that the system is chemically and physically uniform or substantially homogenous throughout or consists of one thermodynamic phase, such a dispersion is called a "solid solution". Solid solutions of abuse-relevant actives are preferred.
The formulation can also comprise one or more additives selected from sugar alcohols or derivatives thereof, maltodextrines; pharmaceutically acceptable surfactants, flow regulators, disintegrants, bulking agents and lubricants. Useful sugar alcohols are exemplified by mannitol, sorbitol, xylitol; useful sugar alcohol derivatives include without limitation isomalt, hydrogenated condensed palatinose and others that are both similar and dissimilar.
Pharmaceutically acceptable surfactants are preferably pharmaceutically acceptable non-ionic surfactant. Incorporation of surfactants is especially preferred for matrices containing poorly water-soluble active ingredients and/or to improve the wettability of the formulation. The surfactant can effectuate an instantaneous emulsification of the active ingredient released from the dosage form and prevent precipitation of the active ingredient in the aqueous fluids of the gastrointestinal tract.
Some additives include polyoxyethylene alkyl ethers, e.g. polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether; polyoxyethylene alkylaryl ethers, e.g. polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether or polyoxyethylene (3) octylphenyl ether; polyethylene glycol fatty acid esters, e.g. PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG-400 dilaurate, PEG-300 distearate or PEG-300 dioleate; alkylene glycol fatty acid mono esters, e.g. propylene glycol mono- and dilaurate (Lauroglycol );sucrose fatty acid esters, e.g. sucrose monostearate, sucrose distearate, sucrose monolaurate or sucrose dilaurate; sorbitan fatty acid mono- and diesters such as sorbitan mono laurate (Span 20), sorbitan monooleate, sorbitan monopalmitate (Span 40), or sorbitan stearate, polyoxyethylene castor oil derivates, e.g. polyoxyethyleneglycerol triricinoleate or polyoxy135 castor oil (Cremophor EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate such as polyethylenglyco140 hydrogenated castor oil (Cremophor RH 40) or polyethylenglycol 60 hydrogenated castor oil (Cremophor RH 60); or block copolymers of ethylene oxide and propylene oxide, also known as polyoxyethylene polyoxypropylene block copolymers or polyoxyethylene polypropyleneglycol such as Pluronic F68, Pluronic F127, Poloxamer 124, Poloxamer 188, Poloxamer 237, Poloxamer 388, or Poloxamer 407 (BASF
Wyandotte Corp.); or mono fatty acid esters of polyoxyethylene (20) sorbitan, e.g.
polyoxyethylene (20) sorbitan monooleate (Tween 80), polyoxyethylene (20) sorbitan monostearate (Tween 60), polyoxyethylene (20) sorbitan monopalmitate (Tween 40), polyoxyethylene (20) sorbitan monolaurate (Tween 20), and the like as well as mixtures of two, three, four, five, or more thereof.
Various other additives may be included in the melt, for example flow regulators such as colloidal silica; lubricants, fillers, disintegrants, plasticizers, stabilizers such as antioxidants, light stabilizers, radical scavengers or stabilizers against microbial attack.
Further, since the acetaminophen-containing overcoat layer has a bitter taste derived from acetaminophen itself, sweeteners and/or flavors etc. may be used as additives to reduce this bitter taste. One preferred way to reduce the bitter taste is a thin additional non-acetaminophen-containing overcoat.
The formulations of the invention can be obtained through any suitable melt process such as by the use of a heated press, and are preferably prepared by melt extrusion. In order to obtain a homogeneous distribution and a sufficient degree of dispersion of the drug, the drug-containing melt can be kept in the heated barrel of a melt extruder during a sufficient residence time.
Melting occurs at the transition into a liquid or rubbery state in which it is possible for one component to be homogeneously embedded in the other. Melting usually involves heating above the softening point of meltable excipients of the formulation, e.g. a cellulose ether/ester, sugar alcohol and/or (meth)acrylic polymer. The preparation of the melt can take place in a variety of ways.
Usually, the melt temperature is in the range of 70 to 250 C, preferably 80 to 180 C, most preferably 100 to 140 C.
When the melt process comprises melt extrusion, the melting and/or mixing can take place in an apparatus customarily used for this purpose. Particularly suitable are extruders or kneaders.
Suitable extruders include single screw extruders, intermeshing screw extruders, and multiscrew extruders, preferably twin screw extruders, which can be co-rotating or counterrotating and are optionally equipped with kneading disks. It will be appreciated that the working temperatures will also be determined by the kind of extruder or the kind of configuration within the extruder that is used. Part of the energy needed to melt, mix and dissolve the components in the extruder can be provided by heating elements. However, the friction and shearing of the material in the extruder may also provide the mixture with a substantial amount of energy and aid in the formation of a homogeneous melt of the components.
In another embodiment, the invention provides an oral, sustained release dosage form characterized in that it has at least two of the following features (a) the abuse relevant drug that is extracted from the formulation by ethanolic solvent, e.g. 40% or 20% aqueous ethanol or both within one hour at 37 C, with or without agitation, is less than or equal 1.5 times the amount of the abuse relevant drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C, (b) the dosage form is resistant to tampering and does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB 501" hardness tester, and (c) the dosage form releases at least 15%, more preferably 18%, and optionally 24% of the drug, but not more than 45%, more preferably 38% and optionally 34% of the drug during the 30 minutes, first hour, or first two hours in in vitro dissolution testing and optionally also in vivo (i.e., in the digestive tract of an animal or human). While not desiring to be bound by any particular theory, it is believed that high initial release rate of acetaminophen from the formulation is accomplished by providing a high drug load in the formulation, especially in the non-core region. Drug loading for a single active ingredient, such as acetaminophen in some embodiments of the inventive formulation can be greater than about 60%, 70%, 75%, 80%, 85%, by weight. The drug loading of acetaminophen can be limited to 80%.
A preferred embodiment of this dosage form is a monolithic form or a solid solution. The term "monolithic" is derived from roots meaning "single" and "stone". A monolithic form or a solid preferably has at least one dimension that is more than 5mm. In monolithic embodiments of the invention, the abuse relevant drug is preferably contained in a single solid, or a single solid solution, element. The monolithic solid or solid solution can optionally be overcoated or combined with other materials. These other materials preferably do not contain a substantial amount of the abuse relevant drug and these materials preferably do not substantially affect the rate of dissolution or dispersion of the abuse relevant drug in vivo or in vitro. The in vitro and/or in vivo release rates of the abuse relevant drug or abuse relevant drugs after about the first hour are preferably substantially constant for at least about 6, 8, 10, 12, or 16 hours. Thus, embodiments of the invention provides a single phase drug formulation that can be adapted to provide a burst of the abuse relevant drug(s) to allow therapeutic levels of the drug to be quickly obtained in the blood of a patient or animal, and to be maintained to provide therapeutic quantities for at least about 8, 12, or 24 hours. Additionally, the drug formulation is preferably suitable for repeated administration to a human or animal once, twice or three times a day.
Advantageously, preferred embodiments of the inventive dosage form release substantially the entire quantity of the abuse relevant drug incorporated into the dosage form.
For example, the inventive dosage form can be adapted to deliver greater than 90%, and preferably 95%, of the drug in in vitro dissolution testing within about 16, and optionally 12 or 9 hours. The cumulative blood concentration, or AUC, cannot be directly known from the time at which 90% of the drug is released from the formulation, however, in general higher AUCs per mg of the abuse relevant drug can be achieved when the drug formulation releases substantially all, or all, of the abuse relevant drug in portions of the digestive tract capable of absorbing the drug into the patient's (or animals) blood system.
In yet another preferred embodiment the invention provides a process for the manufacture of an abuse-resistant drug dosage formulation comprising melt extruding a formulation comprising at least one therapeutic drug further comprising directly shaping the extrudate into a dosage form without (an intermediate) milling step. The melt-extrudate preferably comprises a cellulose derivative, and preferably also comprises a Eudragit polymer. Preferred Eudragit polymers include Eudragit L or Eudragit RS or both, and particularly preferred is Eudragit RL or a combination of Eudragit RL and Eudragit RS.
The melt can range from pasty to viscous. Before allowing the melt to solidify, the melt optionally can be shaped into virtually any desired shape. Conveniently, shaping of the extrudate optionally can be carried out by a calender, preferably with two counter-rotating rollers with mutually matching depressions on their surface. A broad range of tablet forms can be obtained by using rollers with different forms of depressions. Alternatively, the extrudate can be cut into pieces, either before ("hot-cut") or after solidification ("cold-cut") or used in a die injection process. Melt processes involving heated presses optionally can also be calendered.
The formed melt can be optionally overcoated with materials that do not contain substantial amount of the drug with abuse potential. For example, the monolithic dosage form containing the drug of abuse can be overcoated with a color coat, a swallowing aid, or another layer of pharmaceutically acceptable materials. The materials layered over the monolithic form preferably do not materially alter the rate of release of the active ingredient from the dosage form.
In order to facilitate the intake of such a dosage form by a mammal, it is advantageous to give the dosage form an appropriate shape. Large tablets that can be swallowed comfortably are therefore preferably elongated rather than round in shape.
A film coat on the dosage form further contributes to the ease with which it can be swallowed. A
film coat also improves taste and provides an elegant appearance. If desired, the film coat may be an enteric coat. The film coat usually includes a polymeric film-forming material such as hydroxypropyl methylcellulose, hydroxypropylcellulose, and acrylate or methacrylate copolymers. Besides a film-forming polymer, the film-coat may further comprise a plasticizer, e.g. polyethylene glycol, a surfactant, e.g. a Tween type, and optionally a pigment, e.g., titanium dioxide, iron oxides and/or sweeteners or flavors. The film-coating may also comprise talc as an anti-adhesive. The film coat usually accounts for less than about 5% by weight of the dosage form.

EXEMPLARY EMBODIMENTS OF THE INVENTION:
Certain exemplary embodiments of the present invention provide monolithic dosage formulations having biphasic release profile for readily water-soluble drugs having a polymer-containing tablet produced by extrusion and calendering. The formulations preferably have combination of immediate release and controlled release formulations of hydrocodone and acetaminophen compositions. These monolithic dosage formulation, especially having narcotic drugs may have abuse deterrent profiles such that the drug dissolution of the dosage forms has reduced/minimal dose dumping in 40% aqueous ethanol solution. Yet more preferably, these formulations may provide reproducible manufacturing processes offering options for rapid transfer to production scale.
The desired biphasic drug dissolution of acetaminophen can be achieved while retaining a monolithic dosage form by embedding the active ingredient (acetaminophen) in two formulations with differing release rates which are then combined to produce a two-layer or multi-layer tablet. Processes suitable for this purpose include coextrusion methods for the production of multilayer tablets as described in EP 0857062 specifically for extrudate dosage forms. One disadvantage of this technique is that two extruders have to be operated simultaneously and their mass and volume flows have to be coordinated with great exactness.
Especially when shaping the tablet in the calender, the two melts have to be combined with each other in a ratio that is maintained very exactly to ensure compliance with the assay and content uniformity requirements of the tablets as specified in the pharmacopoeias (e.g. USP, Ph. Eur.).
This requires a high level of effort.
It is also possible to manufacture the rapid release acetaminophen portion in a separate tablet which is then incorporated in the still plastic melt of the slow-releasing drug portion during calendering. After cooling, a calendered extruded tablet is obtained which contains a separately embedded rapid release component. Dosage forms of this type are described in US 6,001,391 specifically for extruded dosage forms. One disadvantage of this approach is that the rapid release acetaminophen tablet has to be introduced very precisely into the individual calender cavities to prevent it being completely enveloped by the melt. Only if this rapid release acetaminophen component is located directly at the surface of the tablet can drug dissolution from this separate tablet portion start rapidly enough on contact with aqueous media.
It is also possible to obtain a rapid release acetaminophen component in the tablet by applying a film coating containing acetaminophen. The manufacture of film-coated extruded dosage forms is described in various patent applications. These patent applications do not however, describe a drug-containing film coating designed specifically to achieve biphasic drug dissolution.
The results of the clinical study with an extruded dosage form produced in accordance with the patent applications 11/625,705 and PCT/US07/60864 revealed that about 20% of the acetaminophen contained in the tablet have to be converted to a rapid release formulation to achieve the desired biphasic drug dissolution (for example, > about 30% after 1 h, > about 80% after 8 h). With a total acetaminophen content of about 500 mg per tablet, meant that about 100 mg of acetaminophen had to be rapidly relased.
Applying about 100mg of an active ingredient in a rapid release form onto a tablets is difficult and only possible if certain requirements are fulfilled:
The drug content of the film-coating formulation must be very high so that the layers do not become too thick.

The drug-containing solution or dispersion used for film coating must have a high concentration to avoid long process times which would otherwise make the process uneconomical.
The film coating layer should also offer sufficient mechanical stability even with a large layer thickness, must not be tacky etc. and must be flexible enough that no cracking occurs even with thick layers. Good adhesion on the surface of the extruded cores must also be guaranteed.
The drug dissolution from the film-coating layer should also be rapid when using thick layers (about a maximum of 1 h in a preferred embodiment).
The organoleptic properties of the film-coating layer must also be largely unchanged with large layer thicknesses and during storage for extended periods of time at elevated temperature, high or very low relative humidity or a combination of such (i.e.
no cracking, adhesion, chipping of the coating etc.).
Surprisingly, it has now been found that the above requirements can be fulfilled if finely ground acetaminophen is used for the film coating layers, together with relatively small amounts of a suitable water soluble or water-swellable polymer. It was found that formulations of this type with high active ingredient contents could be achieved, and that the viscosity of the spray solutions was conspicuously low even with very high total solids contents of more than 30% by weight, and that even thick film-coating layers (200 micrometers and more) could be applied in a relatively short time, thereby making the process economical. Drug dissolution was also sufficiently rapid in layers containing above 100 mg acetaminophen.
It was therefore possible to control very precisely the amount of acetaminophen sprayed on and thus also the drug dissolution profile (i.e. release during the first hour) via the layer thickness of the film coating.
Another surprising discovery was that the film coating formulations according to the invention were capable of very effectively smoothing the rough surfaces of the extruded tablets, i.e. the film coating sealed the indentations on the surface of the tablets very effectively. This was surprising considering that almost all commercially available film coatings and the polymers used to produce them actually do not possess and are not intended to possess this very property.
Known polymers and film-coating formulations are designed to reproduce in detail the embossed elements (logos, etc.) and break lines in detail. In other words, "filling in"
of the recesses present particularly in conventionally manufactured tablets is not desired and is to be absolutely avoided (see WO 2006/002808; particular reference is made to this fact in all the samples, see Example 4, page 18: "The embossing was well reproduced, without smearing and bridging effects").
Suitable polymers for the manufacture of the film-coating formulations are water soluble and water-swellable pharmaceutically accepted polymers which have already been used to date for the preparation of film coatings. The basic requirement is that sprayable, preferably purely aqueous solutions or suspensions are produced which have a total solids content (= sum of all the dissolved or suspended constituents including active ingredient) of at least 20% by weight (preferably 25%, particularly preferably 30% or more). The total solids content of the solution or dispersion must also have an active ingredient content of at least 50%
(preferably 60%, particularly preferably 70% or higher). Non-aqueous solutions or suspensions are also possible if non-toxic, pharmaceutically accepted solvents such as ethanol are used.
Mixtures of these organic solvents with water are also possible. In general, however, purely aqueous solutions or suspensions are preferred.
Particularly preferred are polymers which form comparatively low viscosity solutions in aqueous solution even at high concentrations in order to maintain the viscosity of the spray solution within the range in which an acceptable spray behavior of the solution or the suspension is still assured even when using the high total solids contents mentioned above.
Suitable polymers include: non-ionic cellulose polymers such as hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose; cationic polymethacrylates such as Eudragit E, Eudragit NE30D, Eudragit RL, Eudragit RS ; polyvinyl alcohol; polyethylene oxide (high molecular polyethylene glycols with a molecular weight (MW) > 100,000);
polyvinyl alcohol/polyethylene oxide graft copolymers (Kollicoat IR). Preferably, suitable polymers are selected from hydroxypropyl methylcellulose, Eudragit NE30D and polyvinyl alcohol, or combinations thereof. More preferably, suitable polymers are polyvinyl alcohol/polyethylene oxide graft copolymers (e.g.Kollicoat IR, BASF).
The active ingredient (preferred: acetaminophen) must either be soluble in the aforementioned high concentrations in the aqueous or aqueous / organic or purely organic solvents. If (as with acetaminophen) the aqueous solubility is not sufficient, preferably drug suspensions or dispersions can also be used. In this case, however, the decisive factor is that the particle size distribution of the active ingredient should be sufficiently fine since otherwise undesired, i.e. too rapid sedimentation of the suspended active ingredient in the spray solution occurs and/or the spray nozzles of the film coater become blocked. Preferred particle sizes are:
not more than 10%
of the particles above 0.25 mm (particularly preferred: not more than 5%), not more than 20%
(particularly preferred not more than 10%) of the particles above 0.1 mm, and not more than 35% (particularly preferred not more than 20%) of the particles above 0.063 mm. To achieve this finer particle size, the drugs may be comminuted in grinding processes (dry and wet grinding are suitable).
Surprisingly, it was found that the film coating layers according to the invention not only adhere extremely well to the tablets but also do not become brittle or tacky and show no cracking even during storage at elevated temperatures of up to 60 C. There was also no detachment of the coating layer from the tablet core.
Various exemplary embodiments are depicted below. These Examples are being provided for illustrative purposes and they should not be deemed to narrow the scope of the invention.

Example 1: Manufacture of the tablets for film coating A homogeneous powder mixture consisting of 61.8% by weight acetaminophen, 12.6% by weight Eudragit RL, 12.6% by weight xylitol, 6% by weight hydroxypropyl methylcellulose (Methocel K100), 6% by weight hydroxypropyl methylcellulose (Methocel K100M) and 1.0% by weight Aerosil 200 was metered at a rate of 20 kg/h into a co-rotating twin screw extruder (ZSK-40) and extruded at a temperature of about 140 C to produce a homogeneous, white melt ribbon. While still in the plastic state, this melt ribbon was introduced into the roll slit of a counter-rotating forming roller calender, the rollers of which had recesses on their surface from which tablets could be formed directly from the melt ribbon. The resulting tablets had a mean weight of 720 mg after cooling and deburring. The surface of the tablets was rough and uneven in places.
Example 2:
Acetaminophen with a particle size of 13 % greater than 0.25 mm and 68%
greater than 0.063 mm was suspended in water by stirring. The active ingredient settled very rapidly after switching off the stirrer. This suspension was comminuted and homogenized by passing through a colloidal mill. After milling, a solid, powdered polymer (Kollicoat IR, BASF) was added to this suspension (mass ratio acetaminophen/Kollicoat IR = 75:25) to produce a total solids concentration of 30% by weight. Even after adding the polymer the acetaminophen still showed a marked tendency to sedimentation. While continuously stirring this suspension was then sprayed onto the tablets described in example 1 (6 kg) in a film coater (Driam). Samples of tablets were taken after 30, 50, 70 and 90 mg acetaminophen had been applied over the film coat.
In all cases the coating was observed to adhere very well to the tablets, although the surface of the pure white film-coated tablets was still slightly rough due to the still relatively large acetaminophen particles. The loss on drying of the tablets was 1% by weight before and after film coating for all forms.
Film coating process parameters:
6 kg tablet cores Drum speed: 12 rpm Inlet air: 1200 m3/h Inlet air temperature: 65 C
Spraying rate: 40 - 45 g/min Spraying pressure: 4,5 bar Example 3:
Acetaminophen with a particle size of 1% greater than 0.25 mm, 5% greater than 0.1 mm and 16% greater than 0.063 mm was suspended in water by stirring. The active ingredient showed a decreased tendency to settle after switching off the stirrer compared to the material which was used in example 2. Solid, powdered polymer (Kollicoat IR, BASF) was then added to this suspension (mass ratio acetaminophen/Kollicoat IR = 75:25) to produce a total solids concentration of 30% by weight. After adding the polymer, the acetaminophen showed hardly any tendency to settle. This suspension was then sprayed onto tablets (6 kg) which had been produced as described in Example 1 but with a slightly modified tablet geometry, in a film coater (Driam) (process parameters as in Example 2). The tablets were sampled after 30, 50, 70, 90 and 120 mg of acetaminophen had been applied to the film coat. Very good adhesion of the coating on the tablets was observed in all cases. The surface of the pure white film-coated tablets was smooth and uniform.

Example 4: Drug dissolution of the tablets The drug dissolution of the tablets according to Example 1 was determined in an apparatus as per US Pharmacopoeia (USP Dissolution Apparatus II (Paddle), USP XXV; 37 C, 0.01 M HC1, 50 rpm). The amount of active ingredient released from the tablets into the aqueous HC1 medium was determined by HPLC at different intervals.
Tablets without film coat application Drug dissolution measured after 30 minutes: 7%
Drug dissolution measured after 60 minutes: 11 %
Drug dissolution measured after 120 minutes: 17%
Drug dissolution measured after 240 minutes: 27%
Example 5: Drug dissolution of the film-coated tablets The drug dissolution of the tablets according to Example 2 was determined in an apparatus as per US Pharmacopoeia (USP Dissolution Apparatus II (Paddle), USP XXV; 37 C, 0.01 M HC1, 50 rpm). The amount of active ingredient released from the tablets into the aqueous HC1 medium was determined by HPLC at different intervals.

Film-coated tablet with 90 mg acetaminophen in the film coat:
Drug dissolution measured after 30 minutes: 16%
Drug dissolution measured after 60 minutes: 20%
Drug dissolution measured after 120 minutes: 27%
Drug dissolution measured after 240 minutes: 36%

The drug dissolution rates increased by about 10% at each test interval due to the initially rapid release of the active ingredient present in the film coat.

Example 6: Drug dissolution of the film-coated tablets The drug dissolution of the tablets according to Example 3 was determined in an apparatus as per US Pharmacopoeia apparatus (paddle method, USP XXV; 37 C, 0.01 M HC1, 50 rpm). The amount of active ingredient released from the tablets into the aqueous HC1 medium was determined by HPLC at different intervals.

Tablet without film coat application:
Drug dissolution measured after 30 minutes: 7%
Drug dissolution measured after 60 minutes: 12%
Drug dissolution measured after 120 minutes: 19%
Drug dissolution measured after 240 minutes: 29%
Drug dissolution measured after 360 minutes: 37%
Drug dissolution measured after 480 minutes: 43%
Film-coated tablet with 120 mg acetaminophen in the film coat:
Drug dissolution measured after 30 minutes: 28%
Drug dissolution measured after 60 minutes: 35%
Drug dissolution measured after 120 minutes: 43%
Drug dissolution measured after 240 minutes: 53%
Drug dissolution measured after 360 minutes: 62%
Drug dissolution measured after 480 minutes: 69%

The drug dissolution rates increased by about 25% at each test interval due to the rapid initial release of the active ingredient present in the film coat.

Example 7:
The test was performed as for Example 3, but instead of Kollicoat IR a solid trituration based on hydroxypropyl methylcellulose was used which contained a small portion of iron oxide color pigments. Because of the markedly higher viscosity of the aqueous suspension the total solid concentration could only be adjusted to 20% by weight, as a result of which the spraying times increased while the other process parameters remained unchanged. Very good adhesion of the coating on the tablets was observed. The surface of the reddish/brownish film-coated tablets was smooth and uniform.

Example 8:
The test was performed as for Example 3, but instead of Kollicoat IR a solid trituration based on polyvinyl alcohol was used which contained a small portion of titanium dioxide pigments.
Because of the slightly higher viscosity of the aqueous suspension the total solid concentration could only be adjusted to 25% by weight, as a result of which the spraying times increased while the other process parameters remained unchanged. Very good adhesion of the coating on the tablets was observed. The surface of the pure white film-coated tablets was smooth and uniform.
Example 9:
Film tablets manufactured in accordance with Examples 3, 7 and 8 were stored in closed glass bottles at temperatures of 40 C and 60 C. After 1 month no cracks were visible on the tablets and no tackiness was observed. Drug dissolution measured by the method described for Example 4 revealed no changes compared to the values recorded at the beginning of storage.

Example 10:
A film-coated tablet manufactured in accordance with Example 3 (90 mg acetaminophen in the film coating layer) was sampled and a thin section was taken in the transverse direction of the tablet with the aid of a microtome and examined under a microscope. The film coating layer was easily distinguishable from the tablet core in the images. The film coating layer was determined as being about 300 micrometers in the images. The smoothing effect of the coating suspension on the rough tablet surfaces was particularly evident, as also seen in Figures 1, 3 and 4.
Example 11: Dissolution in HC1 and Aqueous Ethanol Following is a description of exemplary methodology for studying rate of dissolution of certain compositions in HC1 and 20% aqueous ethanol. Similar methodology may be used for studying rate of dissolution in 40% aqueous ethanol.
Following apparatus and procedures were use for dissolution in 0.01N
hydrochloric acid and 20/40 % aqueous ethanol:
(I) Dissolution in 0.01 N HC1 Apparatus: USP Dissolution Apparatus II (Paddle) Rotation speed: 50 rpm Media: 0.01 N HC1 Media volume: 900 mL
Temperature: 37 C
Sampling time for 30 h release testing: 30 / 60 / 120 / 180 / 240 / 360 / 420 840 / 1080 / 1320 / 1560 / 1800 minutes Sample volume: 10 mL (no volume replacement) Sample preparation: used as is Analytical finish: UV detection, wavelength 280 nm (II) Dissolution in 20 or 40% Aqueous Ethanol Apparatus: USP Dissolution Apparatus II (Paddle) Rotation speed: 50 rpm Media: 20 or 40% aqueous ethanol Media volume 500 mL
Temperature: 37 C
Sampling time for 30 h release testing: 30 / 60 / 120 / 180 / 240 / 360 / 420 840 / 1080 / 1320 / 1560 / 1800 minutes Sample volume: 10 mL (no volume replacement) Sample preparation: used as is Analytical finish: UV detection, wavelength 280 nm III. Dissolution testin of intact tablets in 0.01 N HC1 at 37 C
a.) Fast releasing formulation (with respect to acetaminophen) in 0.01 N HC1 at 37 C is depicted in Table X. Table IX depicts the composition of the Core and the Overcoat of Formulation 5.

Table IX : Formulation 5:
Core Overcoat 65,42% acetaminophen 150 mg acetaminophen 9,29% Eudragit RL-PO 8 mg Kollicoat IR
9,29% Hypromellose Ph. Eur. USP 2208 Type V 100 (Methocel K100) 9.29% Hydroxypropycellulose Ph. Eur. Type EF
2.99% Polaxamer 188 Ph. Eur./NF
2,8% hydrocodone 1% Aerosi1200 Total weight core: 535 mg Total weight coated tablet: 733 mg Table X depicts dissolution data for hydrocodone (X(a)) and acetaminophen (X(b)).

Table X(a):
[Dru ...............................................................................
............................................
...............................................................................
........................................
...............................................................................
.............................................
...............................................................................
.......................................
:::::::::::::::::::::::::::::::
~~::'::~ ~?o~
~i~~~~~~~
...............................................................................
............................................
...............................................................................
........................................
...............................................................................
.............................................
...............................................................................
.......................................
testing time point (min) mean in %

Table X(b) ...............................................................................
..................................
...............................................................................
.......................................
[Dru:..::~ ~~~~:::ae ~~~~~:
~~
:::
::.............................................................................
..........................................
...............................................................................
.......................................
testing time point (min) mean in %

b.) Slow releasing formulation (with respect to acetaminophen) in 0.01 N HC1 at 37 C is depicted in Table XII. Table XI depicts the composition of the Core and the Overcoat of Formulation 6.

Table XI : Formulation 6:
Core Overcoat 55.88% acetaminophen 120 mg acetaminophen 13.50% Eudragit RL-PO 38.4 mg Kollicoat IR
11.0% Hypromellose Ph. Eur. USP 2208 Type V 100 (Methocel K100) 3.01 % Hypromellose Ph. Eur. 2208 Type V 20000 (Methocel K100M) 13.40% Xylitol Ph. Eur./NF Typ Xylisorb 90 2.21 % hydrocodone 1% Aerosi1200 Ph. Eur./NF
Total weight core: 680 mg Total weight coated tablet: 838.4 mg Dissolution data for hydrocodone (XII(a)) and acetaminophen (XII(b)).
Table XII(a):
[Dru . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . .
...............................................................................
............................................
....................................... ...;.....................
......................................................
~:.:.;.: . . .. .. : :.. . . .
~e~~~i~~?o~~~~~i~~~~~~:aC~. . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . .
...............................................................................
............................................
...............................................................................
........................................
testing time point (min) mean in %

Table XII(b) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . .
...............................................................................
..............................................
...............................................................................
.......................................
:::::::::::...;:;.; : :: ::.:
:a~l:~~n~i~:
::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::>:::>:::>:::>:::::>::
:>:::>
::::::::::::::::::::::::::::::::::::::::::::~:~1:::>::::::>::::::>::::::>::::::
>::::::>::::::>::::::>:::::>::::::>:::
.
~ ::.::.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :..::.: :.: :.: :.: :.::.: :.:
:.: :. . .: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :. : :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
...............................................................................
........................................
...............................................................................
.......................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . .
...............................................................................
..............................................
...............................................................................
.......................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . .
...............................................................................
..............................................
...............................................................................
.......................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . .
testing time point (min) mean in %

IV. Dissolution testing of intact tablets in 40% aqueous ethanol at 37 C
a.) Fast releasing formulation (with respect to acetaminophen) in 40% aqueous ethanol at 37 C
is depicted in Table XIV. Table XIII depicts the composition of the Core and the Overcoat of Formulation 5.

Table XIII : Formulation 5:
Core Overcoat 65,42% acetaminophen 150 mg acetaminophen 9,29% Eudragit RL-PO 18 mg Kollicoat IR
9,29% Hypromellose Ph. Eur. USP 2208 Type V 100 (Methocel K100) 9.29% Hydroxypropycellulose Ph. Eur. Type EF
2.99% Polaxamer 188 Ph. Eur./NF
2,8% hydrocodone 1% Aerosi1200 Total weight core: 535 mg Total weight coated tablet: 733 mg Table XIV depicts dissolution data for hydrocodone (XIV(a)) and acetaminophen (XIV(b)).
Table XIV(a):
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
...............................................................................
............................................
...............................................................................
........................................
; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ;
:. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. : :.
; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :
. .
:.;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;
Dr ::>:::>::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::~ ~:::.: -:: ::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
~:.;.: :: :.; : :: :: :: :.: :.: :: ~;:.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: ::
:: :: :: :: :: :: :: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.
: : :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: :: ::
...............................................................................
.......................................
...............................................................................
........................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
...............................................................................
............................................
...............................................................................
........................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
...............................................................................
............................................
...............................................................................
........................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
testing time point (min) mean in %

Table XIV(b) ...............................................................................
..........................
...............................................................................
...........................................................
...............................................................................
..........................
...............................................................................
..........................................................
;:.;:.;.:.:.......... . :.;:.;.;.;.;.;.;.;.;.;.;.;.;.;.;.;:.;:.;
;..;.;.;.;.;.;.;.;.;.;:.:.;.;:.;
~- :.::. .. . :. . . :::::::::::::::::::::::::.
tAM~ : ~. n::::::>::>::>::::> ::::::::::::::::::::::::::::::>~E::::>;
~. .::::::::::::::::::::::: ::::::::::::.! ...:::::::
~:::::::::::::::::::::::::::::::::::::::::::::::::::::::.
~
................................................~..............................
.........
...............................................................................
..........................................................
...............................................................................
..........................
...............................................................................
..........................................................
testing time point (min) mean in %

b.) Slow releasing formulation (with respect to acetaminophen) in 40% aqueous ethanol at 37 C
is depicted in Table XVI. Table XV depicts the composition of the Core and the Overcoat of Formulation 8.

Table XV : formulation 8:
Core Overcoat 55.88% acetaminophen 120 mg acetaminophen 13.50% Eudragit RL-PO 38.4 mg Kollicoat IR
11.0% Hypromellose Ph. Eur. USP 2208 Type V 100 (Methocel K100) 3.01 % Hypromellose Ph. Eur. 2208 Type V 20000 (Methocel K100M) 13.40% Xylitol Ph. Eur./NF Typ Xylisorb 90 2.21 % hydrocodone 1% Aerosi1200 Ph. Eur./NF
Total weight core: 680 mg Total weight coated tablet: 838.4 mg Table XVI depicts dissolution data for hydrocodone (XVI(a)) and acetaminophen (XVI(b)).
Table XVI(a):
...............................................................................
....................................................
...............................................................................
.................................
...............................................................................
....................................................
...............................................................................
................................
.;.;:.;:~:.;:.;:.; ::::.; . . ::::.... . ;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;::
;
~ ::.;:. .: . .; . . :..
::~
~~::'::~~?o~~::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::::::>:::
:>::::>::::>::::>::::::::::>::::::::>::::::::>::::::::>::::::>::::::>:::>:::~~>
::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::
.:::::::::::::::::::::::: ~ :::
.::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::
...............................................................................
...............................................................................
...............................................................................
......
...............................................................................
....................................................
...............................................................................
................................
...............................................................................
....................................................
...............................................................................
.................................
...............................................................................
....................................................
...............................................................................
................................
...............................................................................
....................................................
...............................................................................
.................................
...............................................................................
....................................................
...............................................................................
................................
testing time point (min) mean in %

Table XVI(b) ...............................................................................
.....................................................
...............................................................................
...............................
...............................................................................
......................................................
...............................................................................
...............................
: ; : ; : ; . ; . ; . ; . ; . ; . ; . ; . ; . ; . ; . ; . ; . ; . ; . ; . ; .
; .
;:;:
~la~n~io : :>:::>:::>:::>:::>:::>:::>:::>:::>:::>::::>::: ;:
>:::>:::>:::>::::::>::::::>:::::>::::::>:::4:>:>:>:
~ . ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; :. ; ; : ; : ; : ; :. ....
:. ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ;
........................................ ; :. ; :. ;
............................. ; :. ; ..... ; :. ; :. ; :. ; . ; . ; :. ; . ; .
; . ; :. ; . ; . ; ; . ; . ; . ;
......
...............................................................................
...............................
..............................................
...............................................................................
...............................
testing time point (min) mean in %

V. Dissolution testing of ground tablets (coffee grinder 60 sec) in 40%
aqueous ethanol at 37 C
In a household coffee grinder 3 extrudate tablet were milled for 60 sec at -20,000-50,000 rpm.
The powder was collected and the to one tablet equivalent amount of powder was transferred to a dissolution vessel for release testing.

To determine the particle size analysis of the sample the powder was collected and sieved through a sieve with a mesh size of 355 m. The material that went through the sieve was sieved again through a sieve with a mesh size of 63 m. The following fractions were obtained:
Fraction 1: particle size > 355 m (- 20 % of the total amount of powder) Fraction 2: particle size > 63 m and < 355 m (- 66 % of the total amount of powder) Fraction 3: particle size < 63 m (-14 % of the total amount of powder) a.) Fast releasing formulation (with respect to acetaminophen) in 40% aqueous ethanol at 37 C
is depicted in Table XVII. Dissolution data for hydrocodone (XVII(a)) and acetaminophen (XVII(b)) are depicted below:
Table XVII(a):
...............................................................................
...................................................
...............................................................................
.................................
...............................................................................
...................................................
...............................................................................
.................................
;:: :;:;:;:;:;:;: :.;:;:;:
;:;:;:;:;:;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.
;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:;:
.;:.;:.;:.;:.;:.;..;:.;:.;::
:.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.:::::
D!~u :>re;:. :>:::>:::>:::>:::>:::>:::>:::>
::>:::>:::>:::>:::>:::>:::>:::>::::::>:::~
::>:>:>:>:>:>:>:>:>:>:>:>:>:>:>:>:>:>:
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . .
testing time point (min) mean in %

Table XVII(b):
...............................................................................
....................................................
...............................................................................
.................................
...............................................................................
....................................................
...............................................................................
................................
...............................................................................
....................................................
...............................................................................
.................................
~ >:: . .; :
: :
~~~i: :::;:;:;:;:;~el~~~~:::a~e ~:~t~: :
~teri:::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>:::::
:>::::::>::::::> :::::::
:::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>:t~i::::41~::~`
~: ::l~ ~Q:H:>::>::>::
. ; :.: ~ :. . . . . : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; :
; : ; : ; : ; : ; :
>:>
:..............................................................................
................................................
...............................................................................
................................
testing time point (min) mean in %

b.) Slow releasing formulation (with respect to acetaminophen) in 40% aqueous ethanol at 37 C
is depicted in Table XVIII. Dissolution data for hydrocodone (XVIII(a)) and acetaminophen (XVIII(b)) are depicted below:
Table XVIII(a):
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
...............................................................................
..................................................
...............................................................................
..................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
::::>::::::>::::::>::::::>::::::>::::>::::>::::>::::>::::>::::>::::>::::>::::>:
:::>::::>::::>::::>:::::::>::::>::::>:::>::>::>::>::>::>::::>::::>::::
>::>::>:
>:::~Il::;: .. .> > : :. . : . .
.:>::>::>::>::>::>::>::>:>:>:>:>:>:>:>:>:>:>:>:
::::::::::::::.::.::::::::::::::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.:
:.::.::.::.::.::.::.::.::.::.::.::.::.
::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.:
:.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::
.::.::.::.:.
:
: :
...............................................................................
.............................................
...............................................................................
..................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
...............................................................................
..................................................
...............................................................................
..................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
...............................................................................
.................................................
...............................................................................
..................................
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . .
testing time point (min) mean in %

Table XVIII(b):
...............................................................................
.........................................................
...............................................................................
...........................
...............................................................................
..........................................................
...............................................................................
...........................
: ::~~~ ~~~~::::~~~t~~a~~~:
:::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>:::>:::>:::>:::>:::>:::>
:::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::~~::.~~....~.
. ~ rk~~ ...................................
~::::::::::::::::::::::::::::::::::::::::::::::::: ~
::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::.
.::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::.
...............................................................................
....................................................
...............................................................................
...........................
...............................................................................
..........................................................
...............................................................................
...........................
...............................................................................
.........................................................
...............................................................................
...........................
...............................................................................
..........................................................
...............................................................................
...........................
...............................................................................
.........................................................
...............................................................................
...........................
...............................................................................
..........................................................
...............................................................................
...........................
testing time point (min) mean in %

VI. Dissolution testin of intact tablets in 0.01 N HC1 at 4 C
a.) Fast releasing formulation (with respect to acetaminophen) in 0.01 N HC1 at 4 C is depicted in Table XIX. Dissolution data for hydrocodone (XIX(a)) and acetaminophen (XIX(b)) are depicted below:
Table XIX(a):
...............................................................................
............................................
...............................................................................
.........................................
...............................................................................
...........................................
...............................................................................
.........................................
. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ;
:. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. . ; :. ;
:. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; :. ; : ;
: ; :. : ; : t : ; : ; : . ; :: .: . . . .. . . . . . : . .. : ; : ; : ; : ; :
; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; : ; :
Dr U: ::~~a e: :. .:~
::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::
::>::::::>::
::::::>::::::>:::::>:::>:::>:::>:::::>::::::>::::::>:::>:::::~1:~:~:::N::~:~I::
:>:::>:::>:::>:::>:::>:::>:::>::::::>::::::>::::
:: :: :.; .: :.: :.: :.: :.: :.: :.: :.;: :.;:.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: .: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :
...............................................................................
......................................
...............................................................................
.........................................
...............................................................................
............................................
...............................................................................
.........................................
...............................................................................
...........................................
...............................................................................
.........................................
...............................................................................
............................................
...............................................................................
.........................................
...............................................................................
...........................................
...............................................................................
.........................................
...............................................................................
............................................
...............................................................................
.........................................
testing time point (min) mean in %

Table XIX(b):
...............................................................................
.............................................
...............................................................................
.......................................
..............................................................................
:.: :.; .: .;: :.;.
: ::~~~ :>:::>:::>:::>:::>:::>
:::>:::>::>::::>::::::>::::::>::::::>::::::>::::::>::::::>::~ :::~~ : ~::::~
~:::::::::::::::::::::::::::::::::::::::::::::::::
~::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::.
...............................................................................
........................................
...............................................................................
.......................................
...............................................................................
.............................................
...............................................................................
........................................
...............................................................................
.............................................
...............................................................................
.......................................
...............................................................................
.............................................
...............................................................................
........................................
...............................................................................
.............................................
...............................................................................
.......................................
...............................................................................
.............................................
...............................................................................
........................................
testing time point (min) mean in %

b.) Slow releasing formulation (with respect to acetaminophen) in 0.01 N HC1 at 4 C is depicted in Table XX. Dissolution data for hydrocodone (XX(a)) and acetaminophen (XX(b)) are depicted below:
Table XX(a):
...............................................................................
............................................
...............................................................................
.........................................
...............................................................................
...........................................
...............................................................................
.........................................
...............................................................................
............................................
...............................................................................
.........................................
.....i..::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>:::
:::::>::::::::>
::::::::::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::
:>::::>:O:::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::
>:::?
:::::::::::::::;:.:::.;:.;:.;::::::::::::::::::::::::::::::::::::::::::::::::::
:::::
::::::::::::::::::::::::::::::::::::::::::::::::.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.
;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:
...............................................................................
......................................
...............................................................................
.........................................
...............................................................................
............................................
...............................................................................
.........................................
...............................................................................
...........................................
...............................................................................
.........................................
...............................................................................
............................................
...............................................................................
.........................................
...............................................................................
...........................................
...............................................................................
.........................................
...............................................................................
............................................
...............................................................................
.........................................
testing time point (min) mean in %

Table XX(b):
...............................................................................
.............................................
...............................................................................
.......................................
..............................................................................
:.; .: .;: :.;.
: ::~~~ :>:::>:::>:::>:::>:::>
:::>:::>::>::::>::::::>::::::>::::::>::::::>::::::>::::::>::~ ::~~ ~::::~
~:::::::::::::::::::::::::::::::::::::::::::::::::
~::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::.
...............................................................................
........................................
...............................................................................
.......................................
...............................................................................
.............................................
...............................................................................
........................................
...............................................................................
.............................................
...............................................................................
.......................................
...............................................................................
.............................................
...............................................................................
........................................
...............................................................................
.............................................
...............................................................................
.......................................
...............................................................................
.............................................
...............................................................................
........................................
testing time point (min) mean in %

VIII. Surface rou _4hness Coating of the extrudated tablets resulted in significant smoothing of the tablet surface as can be seen in Figure 1:
To determine the change in surface roughness coated and uncoated tablets were cut in half along the minor axis. The surface of this cross section was milled to obtain a plain and smooth surface.
Optical micrographs of the cross section were used to determine the average surface roughness.
For analysis, Centre Line Average approach (CLA), was used as depicted in Figure 2, in which the average height per unit length off the centre line is determined. The centre line was put in the micrograph such that the area above and below the line are approximately equal.
The CLA is calculated by using samples at evenly spaced positions according to the following equation:

4. *. r $.

The total length 1 was 4.69 mm, the distance between the increments was 68 m.
For uncoated formulation CLA = 0.56, when (N = 69), as shown in Figure 3.
Whereas for a coated formulation CLA = 0.15, when (N = 69), as shown in Figure 4.

IX. Dissolution testin4 of intact tablets in 0.01 N HC1 at 37 C for different coatin4 thickness a.) Slow releasing formulation (with respect to acetaminophen) in 0.01 N HC1 at 37 C is depicted for various Formulations 9-12 in Tables XXII and XXIII. Compositions of the Formulations are depicted in Table XXI.

..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
. :::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::
~:
~~.:>::>::>::::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>:::>::>::
>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::::>::>::>::>::>:
:>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>:::::>::>::>::>::>::>::>::>::
>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>
::>
.................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
.............................................. .. ............
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
.;:.;.;.;.;.;.;.;
uR:::..::::::::::::::::::~?o~~~:~~~~~a~::::~ >:::>:::>:::
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
60% 60% 60%
Composition 60% acetaminophen acetaminophen acetaminophen acetaminophen 12,6%
12,6% Eudragit 12,6% Eudragit Eudragit RL- 12,6% Eudragit RL-PO
RL-PO RL-PO
PO
6,0% 6,0% 6,0%
Hypromellose Hypromellose Ph. Hypromellose Ph.
6,0% Hypromellose Ph. Eur.
Ph. Eur. USP Eur. USP 2208 Eur. USP 2208 2208 Type V Type V Type V SP 2208 Type V
100(Methocel 100(Methocel 100(Methocel 100(Methocel K100) K100) K100) K100) 6,0%
6,0% 6,0%
Hypromellose Hypromellose Ph. Hypromellose Ph. 6,0% Hypromellose Ph. Eur.
Ph. Eur. 2208 Type V Eur. 2208 Type V Eur. 2208 Type V 2208 Type V
20000(Methoc 20000(Methocel 20000(Methocel 20000(Methocel K100M) el K100M) K100M) K100M) 12,6% Xylitol 12,6% Xylitol Ph. 12,6% Xylitol Ph.
Ph. Eur./NF Eur./NF Typ Eur./NF Typ 12,6% Xylitol Ph. Eur./NF
Typ Xylisorb Typ Xylisorb 90 ylisorb 90 ylisorb 90 1,8% 1,8% 1,8%
1,8% hydrocodone hydrocodone hydrocodone hydrocodone 1 % Aerosil 1% Aerosi1200 1% Aerosi1200 200 Ph. 1% Aerosi1200 Ph. Eur./NF
Ph. Eur./NF Ph. Eur./NF
Eur./NF

50,0 mg 85,0 mg Coating 120,0 mg acetaminophen acetaminophen acetaminophen 16,0 mg Kollicoat 27,2 mg Kollicoat 38,39 mg Kollicoat IR
IR IR

Target weight 833 mg 899 mg 945,2 mg 991,39 mg ....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................

>::::::::>::::::::>::::::::>::::::::>::::::::>
:::::::::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>
::::::::>:::
:::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::
>:::::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>:::
:::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::
......
:::::::::::::::::::::::::::::::::.:::::::::::::::::::::::::::::::::::::::::::::
:.
:::::::::::::::::::::::::::::::::::::::::::::.:::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::
~.::::::::::::::::::::: : . .::::::::::::::.:::: . .:::: ::::
`~rrn ~:Ia - *0..n::9::::::FO:~r:~:ul::
~:::0:::::::::FO~r~ul~iY~~~::::1:~::::: :::: :: ::>::::::>::::::>::::::>.~. M
~''t~~ t~~t~ a~ri~ ::
::::::::::>::::::::::>::::::::::>::::::::::>:::::::::::::::::::::::::::::::::::
::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:.;:.;:; :::::::::::::::::::.::.::.: .::.::.::.::.::.::.::.::.::.::.::.::.:
:.: :.: :.: :.: :.: :.: :..: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
testing point mean in % mean in % mean in % mean in %
(min) ....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
..........................................................
...............................................................................
.......................................
:>::>::>:::::>::>::>::>::>::>::>::>::>::>::>::>::>::>::>::::>::>::>::>::>::>::>
::>::>::>::>::>::>::>::>:::>::>::>::>::>::>:::>::::>::::>::::>::::>::::>::::>::
::>::::>::::>::::>::::>::::>::::>::::>::::>::::>::::>

~. . :: ..: ...:: ......::. ::::::::::::::::::::::::::::::::::::::
::::::::::::::::::::::::::::::::::::::::::::::
::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::.
.:......... ......................
...............................................................................
.......
...............................................................................
.......................................
;:;::.:;:;:;:.;:.;: :t:.;:.;:.;:.; .::;: : ;:;:;:;:;:;:;:: ;:;: ._;:;:;:;:: :
.;:: ;: : ;:;:;:;:;:;:;:: ;:;:
c ~ouOrmula~1FOr~un:rm:?M
;.;.;.;.;.;.;.;.;.;.;.;.;.;.;.;
~ ~ii ::::::::::::::::::::::::::::::::::::::::::::::::::::::::
:::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::::::
:::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::
::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::
:::::::::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::::::>::::::
::::>::::::::::>::::::::::>::::::::::>::::::::::>:::::::::::
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
....................................
...............................................................................
.......
...............................................................................
.......................................
..................................... ......................................
..............................................
..............................................
........................................................................
testing point mean in % mean in % mean in % mean in %
(min) X. Dissolution testin of intact tablets without overcoat in 0.01 N HC1 at 37 C
a.) Fast releasing formulation (with respect to acetaminophen) in 0.01 N HC1 at 37 C is depicted in Table XXV. Table XXIV depicts the composition of the Core of Formulation 13.

Table XXV : Formulation 13 Core No Overcoat 65,42% acetaminophen 9,29% Eudragit RL-PO
9,29% Hypromellose Ph. Eur. USP 2208 Type V 100 (Methocel K100) 9.29% Hydroxypropycellulose Ph. Eur. Type EF
2.99% Polaxamer 188 Ph. Eur./NF
2,8% hydrocodone 1% Aerosi1200 Total weight: 535 mg Dissolution data for hydrocodone (XXV(a)) and acetaminophen (XXV(b)) are depicted below:

Table XXV(a):
[Dru ...............................................................................
....................................
...............................................................................
................................................
...............................................................................
....................................
...............................................................................
................................................
.:: .;: : .;:. .. : .. . . . :. :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
~~~i::'::~~?o~~~~~i~a~ ~aO~~1~~~
...............................................................................
....................................
...............................................................................
................................................
...............................................................................
....................................
...............................................................................
................................................
testing time point (min) mean in %

Table XXV(b):
...............................................................................
......................................
...............................................................................
...............................................
...............................................................................
......................................
...............................................................................
..............................................
: :.; .: ::::::::::::::::::::::::::::::::::
:..::U-00:
:~~~a:::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>:::::
:::>::::::>::::::>::::::>::::>:::>::
::::: ~::~>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::::::>::
~ .::::::::::::::::::::::::::::::::::::::::::::::::
~.::::::::::::::::::::::::::::::::::::::::::::::::::.
...............................................................................
................................
...............................................................................
...............................................
testing time point (min) mean in %

b.) Slow releasing formulation (with respect to acetaminophen) in 0.01 N HC1 at 37 C is depicted in Table XXVII. Table XXVI depicts the composition of the Core of Formulation 13.

Table XXVI: Formulation 14 Core No Overcoat 55.88% acetaminophen 13.50% Eudragit RL-PO
11.0% Hypromellose Ph. Eur. USP 2208 Type V 100 (Methocel K100) 3.01 % Hypromellose Ph. Eur. 2208 Type V 20000 (Methocel K100M) 13.40% Xylitol Ph. Eur./NF Typ Xylisorb 90 2.21 % hydrocodone 1% Aerosi1200 Ph. Eur./NF
Total weight: 680 mg Dissolution data for hydrocodone (XXVII(a)) and acetaminophen (XXVII(b)) are depicted below:
Table XXVII(a):
...............................................................................
.....................................
...............................................................................
...............................................
...............................................................................
......................................
...............................................................................
...............................................
...............................................................................
.....................................
...............................................................................
...............................................
D~~iÃ:.' :::~~Ie~~~:::i~::
:d~~~~~I;orn..e:::>::::>:::::::>::::::::>::::::::>::::::::>:::::::>::::::::>:::
:::::>:::::
:::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::
>~:n :::~:C:I::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>::::::::>:::
:::::>:
~ .: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.
::.::.::.::.::.::.::.::.::.::.:: .
:.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::.::
.::.: :.: :.: :.: :.::.::.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
:.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.: :.:
...............................................................................
................................
...............................................................................
...............................................
...............................................................................
.....................................
...............................................................................
...............................................
...............................................................................
......................................
...............................................................................
...............................................
...............................................................................
.....................................
...............................................................................
...............................................
...............................................................................
......................................
...............................................................................
...............................................
...............................................................................
.....................................
...............................................................................
...............................................
testing time point (min) mean in %

Table XXVII(a):
...............................................................................
......................................
...............................................................................
..............................................
[DI*u ...............................................................................
.....................................
...............................................................................
...............................................
...;: . .. :.. .:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:;:.;:.;:.;:.;:.;:.
;:.;:.;:...;:.;::.;:.;.;
.;::.;:.;.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:
:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;:.;...;:.;:;
..;: .: :.:::: :>:~;.
. . " :;:::~ ` : 1. .,~ ~ ` ' :t$~n:~~.
;1::::>::::>::::>::::>::::>::::>::::>::::>::::>::::>::::>::::>::::
.:::: ~::::::::::::.::::::::::.::::::::::::::::::::::
.................................................~.............................
......................
...............................................................................
...............................................
testing time point (min) mean in %

Example 12: Compare Bioavailability of Test Formulations Against Control The objective of the study was to compare the bioavailability of two test formulations 15 and 16 with that of the reference Control table. The study design included single-dose, fasting, open-label, three-period, crossover study in 21 subjects. Regimen A included one tablet of Formulation 15; Regimen B included one tablet of Formulation 16; Regimen C
included one tablet of Control 1. Blood samples were collected at 0, 0.25, 0.5, 0.75, 1, 2, 3, 4, 6, 8, 10, 12, 16, 24, 36 and 48 hours after the dose on Study Day 1. The following Table XXVIII
illustrates compositions of test Formulations 15, 16 and Control 1. See also Figures 5 and 6 for mean hydrocodone and acetaminophen concentrations for Formulations 15, 16 and Control 1.
Formulations 5, 7 and 15 are substantially identical to each other, however they have been numbered differently based on the different numbering of the tests and experiments. Similarly, formulations and 6, 8 and 16 are substantially identical to each other, however they have been numbered differently based on the different numbering of the tests and experiments. Also similarly Controls 1 and 2 are substantially identical to each other, however they have been numbered differently based on the different numbering of the tests and experiments.
In one embodiment of the invention, a preferred dosage form is Formulation 15 since Formulation 15 provides better blending properties than Formulation 16, both for blending of hydrocodone bitartrate pentahemihydrate and HPMC and blending of all components. Further, Formulation 15 blend provides for better flow properties than Formulation 16 into the extruder.
Also Formulation 15 provides better direct shaping property than Formulation 16 since Formulation 15 is less sticky than Formulation 16. Moreover, Formulation 15 is expected to have better abuse deterrence than Formulation 16.

Table XXVIII:

[.'.oinponent Test ForniuLalions Contrull Ain0tuit (mg,)1T a blet Forinulatioil1 5 FiiriYrrulyatioi116 Tablet Core H-vi:h-ocodc+ne Blt<irtt'atv 15 15 1 i:1 Ac4ti1111111 :Y1;?11411 380 3 -5330 T iiblet Over+r.oaI

Ht'(.hocod.i?Y1? BYti1'trat.e -- --Ac4ti1111111 :Y1?li?11 1 N1 5(1 70 Preliminary pharmacokinetic parameters for Formulations 15, 16 and Control 1 are depicted below in Table XXIX:
Table XXIX:

Ph.irinLai:okinetii: P.u,aineters Regiinen Hydrewi+rclcine {N=2{}) Tnk-jf. Ciniz vUC t ='-L?(-'.ud ti ~2 C:Li F
(1i) {iw mL} (i1g"h!inL) {ng*h inL} {h) (L=1i) Firrniulation 15 44 14.0 '105 1 09 6 22 a4 , ~ {1^c~.i~) {1'7? al {11"'.~t~ {15r' ~l tira rtV
(3 3n 6) ....
...............................................................................
......
FoirnLilatioti 16 44 13.U 2 04 2l`!9 593 45 0 (i~ryi~) ~1t?n,~i) (2CK'ia) {'~?}c'i)J {?:"(t) (181'o) 4.8 1_.t~ 211 214 5.i~S 43.5 C'i~nhri~l 1 t6 3r'.ol Acet,nninoplien (N=20) T:rnx ; ina -U TCt AT-?( 'ud tl:~ C'.Li F
(h) 4ir inL) (116xh!inL) (lig*h;inL) [li) (L=h) 0"4 106 -1.~ 98~ 240 Ft~i1'I1Lil.lf14~T11."= r~ r ty rs ~i i{t~i~sl i4t~:iyl 'r%~sl `=.59 Fillnllllat1{111 16 ~ c~, (4, i~} ~i~.rs {_1 O) i.,24i7 0.83 ;. 13 ""'.1 22 4 6.47 ?i (~(]nt1Y]1 1 ) 1)~. ~ 1 . v: ~. C4 r~ n C~
i r~l (~-1:i,) t_(T t,l ~lTr~f r3`~ t_-l r~l ........................
.................................................................
N=18 Preliminary relative bioavailability of Formulations 15 and 16 versus Control 1 is shown below in Table XXX:
Table XXX:

Relatis'e Pioa4'iiilabiliti-Regirnens PK t'entz':11 V<71ue' Point 9{1 ..6 (_"onficleni:e Test fls. Reference F'sar.Ilnet:er Test Reference Estilnate+ Interv-A
Hp1rcEe.oilcFne Forniulatiot 14 v.r t.-,".isntsisl 1 1 i9 g{:) 116Y6 1.105 1.040 -1..1"y Fornnllnti4,n 1 ti v.s. t'ontrol 1 (_'niax 13.'40 12.626 1.049 0.985 -1.11(i Forniulatiort 1~.r. Contiol I AT_?C"t 199.636 206335 0.968 0.919-1.019 Forlntllti tirni 16 is. {_"ontr+?l 1 ATT{_"t 203.905 1 -106.338 0.988 0.937 -1.042 Fornnllnti4,n 1~; v.s. t'olltrol 1 A[J('~ ^_[7-14 i='- 2 1 t} 11i7 0.473 0.926 -1.{!22_ Frxniulation 16vs t.-'csrltxcsl 1 _kT?C~ 'O&S67 21()187 0.994 0.944 -1..046 ACet8nllllGl)llen FcatYnula tion 1~vs. Control 1 7 +:714 ? 19 :i 0.918 0.858 -0.983 Fcxnnllatis,n 16Z.s. t'i,ntxc,11 t.'.~ 2.39 s 2.193 1.092 1.018 -1.1"2.
FcatYrnilatiin 15 vs. f_'ti3ntrc3l 1 ATJCt 20.151ht) (1.94" 0.899 -(-.998 F?rnlulatlorr 161.s. Contiol I AI_?t "t 2 23 6 3 2 17 322 1.029 0.9~5 -1.i)8#i Forntlllkitis>n 1? Z.s'. t:.:ontrr:,l 1 -'kT?C:o, 22.171 11 QS? 1.1,1{1fC
0.944 -1.i}'7 FotYnulatiinli>tis.f";mtrt3ll AU'. Z '_.4S?2 21.957 1.023 0.956 -1.09^
~-rhlc~~ uitlun of Yhe least silunies meanc4 for lc?ryarYYluxT:.
+ Alitilo aiith"i of the kffi~ rn~e ~t~ t nuniis ref~enLrl of tlie lea k cln~i~ ul~~ils f~~ 1o+,,nrithui Based on preliminary data, the two test Formulations 15 and 16 are bioequivalent to Control 1 with respect to both CmaX and AUC,,,. The initial rate of hydrocodone absorption is slightly slower for test formulations 15 and 16 compared to Control 1.

Example 13: In vitro Drug Release Profiles:
The following Formulations 17 and 18, as shown below in Table XXXI were studied for in vitro drug release profiles and this profile was compared with uncoated core VM-1 and Control 2, as shown in Figures 7 (a) and (b).

Table XXXI:

Formulation Formulation ..................Cvmpanent....................:.~ua~lit~r~t~n!da~rd ....................Functian :.....17 (650m~~...... .....1~ (500m~~......<
Tablet Amvunt (mg)fTaalet Bitartrate USP Drug substance 1~ u 15.0 . ~'. w.~)...... ' f~,61e1 ....y .......... ..........
..<
Acetaminophen USP .Drug.substance 3,_D1_ ii 350.0 ............................................................ u 49.7 ragitL-........... NF/Ph. Eur. ......... Carrier polymer and ......... ...~
~19VJ.....................~654 /0~........
controlled release polymer (13.5 Io) (9.3 l0) ................................. ... <.~..~..~...~.~. .~. ~....
............. ............... ...... <..:... <..~.~.~.
...;.. . ~.~.~. ..;
Hypromellose ~EuB, USP/Ph. Eur. Carrier polymer and 74.8 49.7 type V 100 controlled release polymer (11.0%) (9.3".) Hypromellose 22ug, ..................... ......USPIPh Eur .....
.......Carrier polymer aud.........
........20.5.................................................
type V 20000 controlled release polymer ,,,,,,,,,,,,,,,,,,,,,,,,,_____,,,,,,,,,,,,,,,,,,,,,,,,,,,,,__ -__,,,,,,,,, õ
Hydroxypropylcellulose, Ph. Eur. Carrier polymer and - 49.2 type EF controlled release polymer f9 2 ~) :............................................................._:...............
........................<......................................................
..... 4 .. ....... ................................... Xylitol NF/Ph. Eur Release modifier 91.1 -Poloxamer 188 NF1Ph. Eur. Release modifier - 16.0 C-~<o l01 Col'I'oi'dal'sili'con d'ioxide """"" 'N'F1'Ph"'Eur 'Glidant 5'8 5.4 (1.0%) (1.0%) ...............................................................................
........
...............................................................................
............... ..... .... .....
.., .. ........... ..... ..
...
~Film Ca~[in~l Tab.l..et ....
..~.. . 680 mg 535 mg :..............._ ..........................................._,..................................
..... .............................................................
.................................. ....................................
Acetaminophen USP Drug substance 120.0 150.0 ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,õ` -õ,,,,,,,,, õ , ,, ,,, Kollicoatl' IR In-house Film former 38.4 48.0 Purified water .... USPIPh..Eur....... ....... Solventforfilm~
coatiug......... ............~N1A.............................NIA......
:................................................................:.............
..........................:....................................................
.........:...................................:.............. .
................
Coated Tablet Weight 838.4 733.0 ...............................................................................
...............................................................................
........:.....................................................................
:

Example 14: Manufacturing of tablets by melt extrusion, deburring and film-coating:
For each of the examples according to Table XXXII a homogeneous powder blend was prepared containing all ingredients. In the case of examples 14A to 16A a two-step blending was performed in order to ensure a homogeneous distribution of the low-dose API
component (hydrocodon bitartrate 2.5 hydrate) in the final blend. Blending process is described in Table XXXIII. In the case of examples 14A - 16A a total number of 5 powder samples from each final powder blend prior to extrusion were analyzed with respect to content uniformity of hydrocodone bitartrate 2.5. hydrate.

Table XXXII depicts composition of powder blends before extrusion and final extrudate tablet (after melt extrusion and direct shaping). All Ingredients were tested and released as specified according to US Pharmacopoeia (USP, NF) and/or European Pharmacopoeia (Ph.
Eur.).
Table XXXII:

No. Iu;redient Exainple Example Exainple Example 1 Paracetamol Ph. Eur./USP 55.9 65.4 60.0 61.8 (Acetaminophen) 2 Hydrocodon bitartrate 2.5 hydrate 2.2 2.8 1.8 -3 Hypromellose Ph. Eur./USP 2208, 11.0 9.3 6.0 6.0 Type V100 (Type: Methocel K100) 4 Hypromellose Ph. Eur./USP 2208, 3.0 - 6.0 6.0 Type V20000 (Type: Methocel K100M) Ammoniummethacrylat- 13.5 9.3 12.8 12.6 Copolymer (Typ A) Ph. Eur./NF
(Type: Eudragit RL PO) 6 Hydroxypropylcellulose Ph. Eur. - 9.2 - -(Type : Klucel EF) 7 Xylitol Ph. Eur./NF 13.4 12.6 12.6 (Type Xylisorb 90) 8 Poloxamer 188 Ph. Eur./NF - 3.0 - -(Type : Lutrol F68) 9 Colloidal silica P. Eur./NF 1.0 1.0 1.0 1.0 (Type: Aerosil 200) Table XXXIII: Blending process for examples 14 - 17 Ste ) Exam ple 14B Etain ple 1,513 Exam ple 16B Exam lc 17B
1 Blending of #2, #3, Blending of #2, #3, Blending of #2, #3, #4, #9 (according #6, #9 (according #4 (according to to Table XXXII) to Table XXXII) Table XXXII) One-step-blending 2 Adding #1, #5, #7 Adding #1, #5, #8 Adding #1, #5, #7, of all ingredients (according to Table (according to Table #9 (according to according to Table XXXII) to blend XXXII) to blend Table XXXII) to XXXII
from step 1. from step 1. blend from step 1.
3 Blending the whole Blending the whole Blending the whole mixture mixture mixture Total 12 kg 12 kg 3 kg 50 kg batch size The final blend from examples 14B - 7B was dosed in a co-rotating twin-screw extruder at a constant feeding rate. The homogeneous, white drug-containing melt leaving the extruder nozzle was directly shaped into elongated tablets by calendering between two counterrotating rollers having depressions on their surface according to the dimensions listed in Table XXXIV. Process parameter settings of melt extrusion and calendering are listed in Table XXXIV.
Table XXXIV depicts melt extrusion and direct shaping (calendering) process:
Table XXXIV.

Process parameter Example 14C Example 15C Exainple 16C Example 17C
settiug Extruder 18 mm 18 mm 18 mm 40 mm (screw diameter) Tablet dimension 19.0 / 6.9 / 3.0 20.0 / 5.9 / 2.5 17.5 / 7.97 / 7.6 19.0 /
6.9 / 3.0 (calender roller mm mm mm mm depression dimension) (length / width / height) Extrusion temperature 129 C 124 C 140 C 140 C
(melt temperature) Calender temperature 11 C 20 C 11 C 11 C
Extrusion throughput 1.5 kg/h 1.5 kg/h 1.5 kg/h 25 kg/h Batch size 12 kg 12 kg 3 kg 50 kg Tablets according to examples 14C, 15C and 17C were transferred into a Driam 600 film-coater.
In a first step the tablets were tumbled in the coater at maximum rotation speed in order to polish the tablets and to remove the seems surrounding the tablets which derive from the calendering shaping process. This material which was removed from the tablets was removed from the coating drum together with the exhausting air. After this "deburring" step film-coating of the tablets was directly started in the same coater. In the case of example 16C
tablets were placed in closed stainless steel container and tumbled for 10 minutes once removal of edges and seems was complete. Tablets were then dedusted on a sieve and transferred to the same Driam film-coater as in the case of the other examples. Composition of film-coating layer and process parameter settings of deburring step and of subsequent film-coating are listed in Table XXXV.
Table XXXV depicts deburring of tablets after calendering Tab1e XXXV:

Process parameter Example 14D Example 15D Example 16D Exanille 17D
setting Deburring time in 20 min. 94 min. - 60 min.
Driam film-coater Deburring time in - - 10 min. -stainless steel drum Drum temperature 25 C 25 C 25 C 25 C
Tablet weight (mean) 684.3 mg 536.4 mg 840.7 716 mg after deburring Acetaminophen drug 382. 5 mg 350.8 mg 500.4 mg 442.5 mg content per tablet (calculated according to composition and mean tablet weight) Hydrocodone 15.0 mg 15.0 mg 15.1 mg -bitartrate 2.5 hydrate drug content per tablet (calculated according to composition and mean tablet weight) Batch size 4.9 kg 6.5 kg 1 kg 7.8 kg Manufacturing of the film-coating suspension for examples 14E - 16E was generally prepared by the following steps: First, acetaminophen was dispersed in water at room temperature during stirring. To this suspension the polymer (Kollicoat IR) was added and stirring was continued until a homogeneous suspension was formed. This suspension was directly used for film-coating.

Stirring was continued during the whole film-coating process. For examples 14E
- 17E a ready to use acetaminophen powder was used (Rhodia, acetaminophen "fine powder"). No additional sieving or micronizing was performed. Composition of film-coating suspensions are summarized in Table XXXVI.
Table XXXVI depicts composition of film-coating suspension Table XXXVI:

Example 14E Example 15E Example 16E Example 17E
Rel. amount of 22.73 %
acetaminophen Acetaminophen 1 % > 0.25 mm particle size 5 % > 0.1 mm (Rhodia fine 16 % > 0.063 mm powder ) Rel. amount of 7.27 %
polymer (Type:
Kollicoat IR) Rel. amount of water 70.0 %
(purified) Film-coating of the deburred tablets was performed in a Driam 600 film-coater.
Process conditions, parameter settings and data from final film-coated tablets are listed in Table XXXVII. In the case of all examples 14F - 17F samples were taken at different time point during main phase of film-coating. This was to study the influence of different amount of coating layer thickness on drug release of both acetaminophen and hydrocodone bitartrate from the film-coated tablets. Spray rate during main phase of film-coating was at maximum rate of the peristaltic pump dosing the acetaminophen/Kollicoat IR suspension. Higher spray rates should be possible.

Table XXXVII depicts film-coating process conditions Table XXXVII:

Process parameter Example 14F Example 15F Example 16F Examhlc 17F
setting Pre-heating phase inlet air temperature 65 C
spray rate -time 10 min.
Starting Fhasc I
inlet air temperature 65 C
spray rate 16 g/min. 15 g/min. 10 g/min.
time 5 min. 6 min. 9 min.
St'irting rh~isc 2 inlet air temperature 65 C
spray rate 21 g/min. 20 g/min. 25 g/min.
time 10 min. 10 min. 8 min.
Main hhasc inlet air temperature 65 C
spray rate 31-42 g/min. 28-47 g/min. 20-44 g/min. 30-48 g/min.
time 131 min. 230 min. 193 min. 159 min.
Dryincoolin~ Fhase inlet air temperature 25-30 C
spray rate - - - -time 5 min. 5 min. 5 min. 5 min.
Batch size 4.4 kg 6.1 kg I kg 7 kg Dimension of film- 19.46 mm 20.63 mm 19.45 mm 19.53 mm coated tablets (mean) 7.82 mm 7.32 mm 10.66 mm 7.62 mm (length / width / height) 7.07 mm 6.41 mm 7.71 mm 7.23 mm Weight of film-coated 848.2 mg 744.8 mg 1018.4 mg 872 mg tablets (mean) Weight of coating 157.9 mg 208.4 mg 177.7 mg 156 mg layer per tablet (calculated) Acetaminophen drug content per film- 119.6 mg 157.9 mg 134.6 mg 118.2 mg coated tablet in film-coating layer (calculated) Total acetaminophen 502.1 mg 508.7 mg 635 mg 560.7 mg drug content per film-coated tablet (calculated) Total hydrocodone 15.0 mg 15.0 mg 15.1 mg -bitartrate 2.5 hydrate drug content per film-coated tablet (calculated) Generally, certain preferred embodiments of the present invention provide dosage forms and methods for the delivery of drugs, particularly drugs of abuse, characterized by resistance to solvent extraction; tampering, crushing or grinding, and providing an initial burst of release of drug followed by a prolonged period of controllable drug release.

Further, as shown below in Table XXXVIII, in one preferred embodiment, the present invention provides a pharmaceutical composition having a core and a non-core layer, comprising: (a) hydrocodone, a pharmaceutically acceptable salt or a hydrate thereof, and (b) acetaminophen or ibuprofen. In this embodiment, at least 75% all of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core, and the acetaminophen or the ibuprofen is the non-core layer. Further, this composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily. Preferably, greater than 90% of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core. More preferably, substantially all of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core. In another embodiment, the core further comprises acetaminophen or ibuprofen. More preferably, the core further comprises acetaminophen.

+ \O N~~~ O~~ N~ M O~ GO GO ~ N GO GO l~ ~ GO l~ M-- M
M 01 01 M N O N~,-~ N 01 GO ~O O O N~ N O O GO
l~ N N N N l~ 01 M4 -:t -T M N M
4 GO 00 "O -/~-i V~ r" `O ~ 01 O--~ ~ l~ O l~ N 01 01 `O l~ ~ N~O `O l~ ~-- ~ GO M M~ l~
o~r ~~ O M O`O M M M-- 01 N O M 01 01 ~

V G~ ~-- GO ~~`O GO 01 M l~ 01 GO O GO l~ GO GO `O l~ `O ~ 01 GO N-- O 01 ~,~ y N M N N O O GO O~~ O 01 ~'~ N GO O O O GO
N--O~~ GO --~ M-- l~ M GO M N~O O 01 N M N O~ l~ GO -- ~ 01 --~ M
N M OAO ~~~\O -- N--~ --~ O 01 N M M M GO ~ M~~~ M M~
C O~~ N l~ ~ O GO M~~~ l~ ~ N~O O N O~ ~ GO O~ ~~ O l~ O
01 00 ,--~ 00 M M N--~ W" 01 O~
O O O O O ~ N N O c 'n GO O M l~ -- 01 GO `O N O~ M -- l~ M O
GO ~ 01 O O l~ N~,O O" l~

~
~
GO N N l~ ~-- `O O O~~ M-- M~~ N-- l~ O N l~ M l~ N GO M
M M M 01 l~ O O~ ~~~ M~ N O~ ~`p M M M N~
C."

..~
GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~
W GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO
GO GO
r.- b0 b0 b0 b0 b0 b0 b0 b0 b0 =~ ~ ~ ~ ~ ~ ~ ~ ~ ~
~

M
I I I I
' U U U U U
H a a ~ 01 l~ N GO -- p~ M 01 -- ~ 01 AO GO N l~ GO O O 01 M GO 01 01 ~~-- -- GO
N N N N N O~~`"' N N O 01 GO GO O O O l~ 01 GO O O O 01 01 ~~~ O GO l~
p~-- GO N N N~ GO M M`O N~O O`O M~ O O O`O 01 GO M`O M-- GO l~
pp ~ 01 M N-- -- M O~ N M O l~ O M N GO AO ~-- M N 01 GO --~ l~ GO
N N~ 01 GO O O~ 01 ~`O ~p O~`O N N N O~ GO M M~
N N N,--i N O M~" N N O GO `O l~ O O O `O GO l~ O O O GO GO 01 O~ O l~ 01 -- GO M p~~ N M -- GO N GO -- ~ l~ ~ 01 ~ GO AO O 01 N~ N O O~ l~
01 GO O ~O `O W) W) 01 M -t M O M `O M ~t kn 00 `O
N-- -- p N M -- -- O~~~ O O O ~\O O O O AO AO l~ O O O~ l~
\O 01 N N M ~ ~ \O M --~ 01 GO ~O GO GO N --~ 01 AO M l~ N N O ~p p-ti N l~ \O 01 ~~ M l~ \O 01 l~ GO AO p M-ti l~ 01 AO M~~ GO O~ N`"
01 l~ ~ l~ 01 N-- GO 01 ~`O M N~~ l~ N 01 M~`O N l~ 01 M O~~~
N~ M~ M N~ N~-- M N 01 O 01 N M N N~ O N M `O 00 M~~'t~~
~~~ N GO O~ p~ ~ N GO O~ O GO O~ GO N N O~~ GO N~ O`O 01 O l~ 01 O GO AO 01 O 00 01 M N ~O N
O O O`"' N O O O N N~ O O O N N N O O O ~~~ O O O~ N

~
GO GO 01 p p" N N O~ N~O ~\O ~ l~ O~~~\O GO 01 ~ l~ ~~ l~
O O N - 01 `O GO -~ `O o0 01 -t kn -t 01 GO -t W) -t W) M o0 01 l~
N N N-- N O N~ ~ N O`O ~`O p p p~ p p p l~ l~ GO O O O`O GO
GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~

`O M N 01 GO GO `O l~ `O ~ M N N~ GO GO l~ N N O N~O M GO N~~ N O N~
~ O N M O O O O O O AO AO N N N N O ~ M ~ ~ ~
l~ N~O 01 M l~ l~ O l~ l~ 01 N~O ~~ O GO O l~ ~~~ ~~~~ N GO ~
~~~ O~~~~~~ O O O O O O O O AO 01 M~ N N 01 01 M~\O N~
O O O ~ 01 01 ~~ O ~~~ O O O O O O M N~ ~~~ N N 01 01 01 ~ O O
O~ M GO l~ 01 ~`O M 01 O~~~~ N~ N l~ ~ GO `O ~ l~ M
. O~~ l~ l~ `O l~ GO N N N 01 ~~~ M N~ N GO 01 ~~~--~ --~ O O O O N ~`O M~`O 01 M
M N-- GO AO N~ GO -- -- p1 GO GO -- M\O ~~~ OAO l~ \O 01 01 AO 01 l~ GO M~
l~ GO `O GO O l~ O `O

~ M~ O O M 01 O M`O --~ M~~~~~~ `O l~ O GO GO O O
N~O 01 O 01 N 01 01 GO O\O 01 M~ N~-~ p~ -~ -~ p O~ O GO O M N~ N~O N~~~~ O O
-- -- -- -- -- -- M ~-- -- N -- O O O O O O l~ l~ - N -- N M
01 ~ `O N GO
O O O GO l~ 00 -~ -- p 01 GO GO O O O O O O N N~~~-~ N N l~ l~ O 01 O O
O O O O O O

01 O O 01 M M~~ N~~ N O N N
N N M N M M O O O O O O ,~ = O O O ' '~ M~ M
O O O O O O O O O O O O `~ O O O

~
`O GO N GO GO `O M N~~ M l~ GO 01 ~ 01 -- GO l~ `O M
GO 01 O~ GO 01 O M`O `O O 01 01 01 01 M 01 N~ N M M O O O O O O N M~
O O O O O O

GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO --~ GO GO
V V V ~ H

M -- 01 N + N M -- GO -- ~ GO \O 0 M -- \O 01 M O
`O GO M y~õ GO 01 M~~ 01 `O `O N--~ 01 01 l~
M I:t O O O O 01 N -- -- -- 4 M M \O 4 4 N GO ~"" --~ M 01 ~ ~ ~ `O GO `O M N ~O M `O N N l~ 01 ~ M
--~ 01 01 --~ A O O M~ 0~ l~ GO M GO AO 01 O M~ 01 ~ 01 01 ~~--~ AO
N N N M N N O O O O ~~~ O O O N~~~ o0 01 M M N
~
O O O o~r y-- -- ~ M GO `O l~ `O M~ M~ 01 O GO l~ l~ ~~ GO --~
M N M
o0 N -- -- -- 01 -:t N N N W') 4 4 l~ l~ \O 01 4 l~ kn N ~ \O O GO O 01 N 01 ~ N ~O ~
O O O O D4 01 01 N M N~~ O N~ M~ l~ O~~~~~ M
O O O O ~`O N~~~ N~~~ O O N l~ N O~ N~ 01 O`O
\O GO AO AO ~O " O O O 01 m M N N N m oc O O O O GO `O N 0 0~~ M O l~ `O GO 01 ~ O l~ M l~ O 01 GO
N N N N z -- 0 O O O ~~~ O O O \O N~~~ GO N N N
`O M N l~ `O O`O ~~~`O -- ~ N~ N O O`O `O l~ N N M~ O M
~~~~ O~ O O O O `~ N O O O O N M~ O O O N~~ O~
O

GO GO l~ ~" M ~ _M GO N 01 O AO 01 ~ N --~ 01 M ~ \O
M M l~ ~~~.c N N N~ M O N O OM4 O O`O
C."

~~~~ ~ GO GO --i GO GO --i GO GO --i GO GO --i GO GO --i GO GO --i GO GO --i GO GO
~~~~ Sr y GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO GO
~ O O O O

-- 01 AO O :t W') M l~ - AO 01 - :t W) N l~ W) 01 :t `O 4 O~~~O O GO 01 ~~~ O O 01 01 01 M~ M 01 `O O~
\O ~ l~ N N M -- -- p p -- -- -- -- -- -- M ~ -- -- -- -- M -- -- -- -- M M

01 `O N ~ l~ 01 ~ ~ O `O 01 N O ~ 01 N O N GO 01 GO `O ~
01 GO GO M~~ `O `O `O `O M~ O 01 01 01 O O--~ 01 01 `O GO
. N ~O GO GO 01 N M M N ~~~ N N O O O O O O O O O O N N~ O O O ~ N~ ~ O O ~~
OAO O~ l~ ~ O GO AO M O~ GO M~~ N GO O M GO AO ~~~ 01 O O
--~ ' ' ' 01 GO 01 GO O N O 01 01 O~ M M M N O ~~~ --~
= N --~ M GO
\O N N N M -- O O O -- -- -- O O -- M -- -- -- -- M M -- -- -- -- N M
M~ O l~ O l~ ~~ N~~~O ~ O GO ~ N O~ `O M~ O`O ~~`O 01 l~ `O O~ 01 M ' N~ OAO ~\O 01 l~ l~ --~ N--~ 01 O M\O N N--~ --~ O N
. O O l~ O N
M~~ M N N N M M~ O O O O O O O O O N M~ O~ N N~ ~~~ N N
GO N l~ l~ O~ l~ O~ M GO ~ GO `O 01 ~ O`O N O O GO GO GO l~ N M N N
~,--~ GO GO M~~~ N GO M--~ M N~ N`O GO N`O 01 ~--~ GO M GO GO 01 ~-- O N M 01 GO l~ N GO M~ l~ O--~ GO 01 GO GO M N GO l~ M~--~ l~ M l~
`O M `O M -- ~ `O ~ `O O 01 l~ `O GO M `O GO 01 GO l~ --N N N N ~~~ N N O O O O O O O O O O ~ N O O O O ~~ O O O O ~~
M~~ . GO M M M M N N M N--~ N N l~ N~ M~ N --~ M M N GO
O~~ N ~~~ O~ O O O O O O O O O O O~ O O O O O~ O O O O O O

N~~~O 01 01 N 01 -- M~ O l~ GO GO O M--~ O N--~ 01 ~\O M GO --~ N--~ M\O l~ 01 ' '~ N N `O l~ GO O 01 GO GO 01 --~ N--~ --~ ~O N~~ M N M`O
--~ GO GO --i GO GO --i GO GO --i GO GO --i GO GO --i GO GO --i GO GO --i GO
GO --i GO GO --i GO GO --i GO GO

`" A e A A A
V V V V V

-- l~ O:t 01 M- W) N~ M 01 M M-- O
I-O GO `O l~ ~~~ GO GO GO
N O O N O O O M~ N~~
. . . . . . . . . . . . . . . .

~--~ ~--~ ~--~ ~--i -- N -- -- -- -- ~ -- -- --N

~--~ 01 01 GO GO AO O \O ~--~ GO 01 M~--~
-- M M\O l~ ~--~ l~ O~--~ AO AO AO l~ M\O l~

O~ O O O O O O O O N M O O O

01 `O GO 01 O~~ M O M
N N N~`O N N M O~~ N M M
O O O O O O O O O O~~ O O O O
N

\O 01 O O~~ O GO M~\O AO GO l~

V V V
x x x tb tb tb tb tb tb tb tb tb tb ~

~
~

In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose. In another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose. In yet another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6 ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose. Other embodiments of the dosage form include about 5-20 mg of hydrocodone bitartrate pentahemihydrate and about 400-600 mg of acetaminophen. Yet another embodiment of the dosage form includes 10-15 mg of hydrocodone bitartrate pentahemihydrate and about 500-600 mg of acetaminophen.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. When administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg. In another embodiment, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg. In yet another embodiment, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
Preferably in this embodiment, the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone bitartrate pentahemihydrate.

In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone of about 0.18 ng/mL/mg to about 1.51 ng/mL/mg, and a plasma concentration at 1 hour Cl for acetaminophen of about 2.34 ng/mL/mg to about 7.24 ng/mL/mg.
In preferred embodiments such as Formulation 15, the dosage form produces a C
l for hydrocodone of about 0.32 ng/mL/mg to about 1.51 ng/mL/mg and a Cl for acetaminophen of about 2.34 ng/mL/mg to about 5.50 ng/mL/mg.
In certain other embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting.
Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone from about 0.30 ng/mL/mg to about 1.06 ng/mL/mg, and a C l for acetaminophen from about 2.75 ng/mL/mg to about 5.57 ng/mL/mg. In preferred embodiments, the dosage from produces a C l for hydrocodone from about 0.45 ng/mL/mg to about 1.06 ng/mL/mg and a C l for acetaminophen from about 2.75 ng/mL/mg to about 4.43 ng/mL/mg.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 3.63 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 2.76 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.79 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.23 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.

In preferred embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen of 1.80 0.42 g/mL with the 95% confidence interval for the mean value falling between about 1.61 g/mL to about 2.00 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. The 95%
confidence interval of combined Cl for hydrocodone and acetaminophen for the preferred embodiments and the Control overlapped. The 95% confidence interval for the mean value of combined Cl for hydrocodone and acetaminophen for the Control ranged from about 1.46 to 1.96 g/mL, after administered as a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen to the human patient. The Control provides sufficient plasma levels of opioid and nonopioid analgesic to provide a reduction in pain intensity within about 1 hour after administration.
When administered to a population of healthy North Americans or Western Europeans, particularly when the formulation is adapted to be suitable for, or intended for, administration to a human every 12 hours as needed, about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C. In another embodiment, about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C.
Further, in another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60%
to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours. In another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and at least 90%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours. In another embodiment, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and at least 95%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours. Yet in another embodiment, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours. In another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 1 lhours to about 12 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours. In yet another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
However, when the a slow-release version of the formulation is adapted to be suitable for, or intended for administration to a human, twice daily, as needed, then at least 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours. In another embodiment of the slow release formulation, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours. In another embodiment of the slow release formulation, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours. In another embodiment of this slow release embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours. In yet another embodiment of the slow release formulation, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
In a preferred embodiment, the present invention provides a composition where the core layer comprises an excipient or a mixture of excipients capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug.
Further, in a preferred embodiment, the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer.
Most preferably, the composition comprises about 500mg of acetaminophen and about 15 mg of hydrocodone bitartrate pentahemihydrate. In another embodiment, the non-core layer, or the tablet layering may be prepared by another methodology. In this methodology the film-coating layer is separately manufactured by extrusion and the extrudate is shaped into a foil. This foil is introduced into the calendar during manufacturing of the cores. This method is especially suitable for thick layers (saving long spray-coating time) and is a solvent-free process. This technology is also known as the Xellex technology.
In another exemplary embodiment, the present invention provides a pharmaceutical composition having a core and a non-core layer, comprising: (a) an abuse-relevant drug, a pharmaceutically acceptable salt or a hydrate thereof and a non-abuse-relevant drug or a pharmaceutically acceptable salt thereof in the core layer, and (b) a non-abuse-relevant drug, a pharmaceutically acceptable salt or a hydrate thereof in the non-core layer. Preferably, this composition is characterized by at least one of the following features:
i) the amount of abuse-relevant drug that is extracted from the composition by 40% aqueous ethanol within one hour at 37 C in vitro is less than or equal 1.5 times the amount of the abuse-1 5 relevant drug that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the abuse-relevant drug and not more than 45% of the abuse-relevant drug during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the non-abuse relevant drug within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the non-abuse relevant drug and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the abuse-relevant drug upon grinding increases by less than 2-to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37 C , vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0. 1 to about 0.2.
In this composition, the amount of the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 C is about 70% to about 130% of the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C. In another embodiment, the amount of the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 C is about 70% to about 90% of the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C.
In yet another embodiment, the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 C is about 75% to about 90% of the amount of the drug that is extracted by 0.01 N hydrochloric acid within one hour at 37 C.
Another embodiment of the present invention provides a pharmaceutical composition having a core layer and a non-core layer. In this composition the core layer comprises a mixture of: (a) at least one opioid; and (b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof. The non-core layer comprises at least one non-opioid analgesic. Further, these compositions are adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily. Preferably, the core layer further comprises at least one non-opioid analgesic. In a preferred embodiment, the composition is characterized by at least one of the following features:
i) the amount of abuse-relevant drug that is extracted from the composition by 40% aqueous ethanol within one hour at 37 C in vitro is less than or equal 1.5 times the amount of the abuse-relevant drug that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the abuse-relevant drug and not more than 45% of the abuse-relevant drug during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the non-abuse relevant drug within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the non-abuse relevant drug and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the abuse-relevant drug upon grinding increases by less than 2-to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37 C , vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0.1 to about 0.2.
In one embodiment, the opioid is selected from the group consisting of alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levophenacylmorphan, levorphanol, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbulphine, narceine, nicomorphine, norpipanone, opium, oxycodone, oxymorphone, papvretum, pentazocine, phenadoxone, phenazocine, phenomorphan, phenoperidine, piminodine, propiram, propoxyphene, sufentanil, tilidine, and tramadol, and salts, hydrates and mixtures thereof.
Further, the non-opioid analgesic is selected from the group consisting of acetaminophen, aspirin, fentaynl, ibuprofen, indomethacin, ketorolac, naproxen, phenacetin, piroxicam, sufentanyl, sunlindac, interferon alpha, and salts, hydrates and mixtures thereof. Preferably, the opioid is hydrocodone and the non-opioid analgesic is acetaminophen or ibuprofen. More preferably, the opioid is hydrocodone and the non-opioid analgesic is acetaminophen.

In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone of about 0.18 ng/mL/mg to about 1.51 ng/mL/mg, and a plasma concentration at 1 hour Cl for acetaminophen of about 2.34 ng/mL/mg to about 7.24 ng/mL/mg.
In preferred embodiments such as Formulation 15, the dosage form produces a C
l for hydrocodone of about 0.32 ng/mL/mg to about 1.51 ng/mL/mg and a Cl for acetaminophen of about 2.34 ng/mL/mg to about 5.50 ng/mL/mg.
In certain other embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting.
Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone from about 0.30 ng/mL/mg to about 1.06 ng/mL/mg, and a C l for acetaminophen from about 2.75 ng/mL/mg to about 5.57 ng/mL/mg. In preferred embodiments, the dosage from produces a C l for hydrocodone from about 0.45 ng/mL/mg to about 1.06 ng/mL/mg and a C l for acetaminophen from about 2.75 ng/mL/mg to about 4.43 ng/mL/mg.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 3.63 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 2.76 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.79 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.23 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.

In preferred embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen of 1.80 0.42 g/mL with the 95% confidence interval for the mean value falling between about 1.61 g/mL to about 2.00 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. The 95%
confidence interval of combined Cl for hydrocodone and acetaminophen for the preferred embodiments and the Control overlapped. The 95% confidence interval for the mean value of combined Cl for hydrocodone and acetaminophen for the Control ranged from about 1.46 to 1.96 g/mL, after administered as a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen to the human patient. The Control provides sufficient plasma levels of opioid and nonopioid analgesic to provide a reduction in pain intensity within about 1 hour after administration.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose. In another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose. In yet another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6 ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. When administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg. In another embodiment, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg. In yet another embodiment, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
Preferably in this embodiment, the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone.
When administered to a population of healthy North Americans or Western Europeans, particularly when the formulation is adapted to be suitable for, or intended for, administration to a human every 12 hours as needed, about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C. In another embodiment, about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C.
Further, in another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60%
to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours. In another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and at least 90%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours. In another embodiment, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and at least 95%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours. Yet in another embodiment, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours. In another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 1 lhours to about 12 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours. In yet another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
However, when the a slow-release version of the formulation is adapted to be suitable for, or intended for administration to a human, twice daily, as needed, then at least 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours. In another embodiment of the slow release formulation, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours. In another embodiment of the slow release formulation, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours. In another embodiment of this slow release embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours. In yet another embodiment of the slow release formulation, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
In a preferred embodiment, the present invention provides a composition where the core layer comprises an excipient capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug. Further, in a preferred embodiment, the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer. Most preferably, the composition comprises about 500mg of acetaminophen and about 15 mg of hydrocodone bitartrate pentahemihydrate.
In another embodiment, the present invention provides a pharmaceutical composition having a core layer and a non-core layer. In this composition, the core layer comprises a mixture of (a) at least one opioid and at least one first non-opioid analgesic; (b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof. The non-core layer comprises at least one second non-opioid analgesic. Further, the composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily. In this embodiment, preferably, the opioid comprises hydrocodone and the first and the second non-opioid analgesic comprises acetaminophen or ibuprofen. More preferably, the opioid comprises hydrocodone and the first and the second non-opioid analgesic comprises acetaminophen.
Further, in this embodiment, the non-core layer comprises: (a) acetaminophen; and (b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof.
Preferably, the polymer or copolymer is selected from the group consisting of: hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose; polymethacrylate, polyvinyl alcohol, polyethylene oxide, and combinations thereof. More preferably, the polymer or copolymer is selected from the group consisting of: hydroxypropyl methylcellulose, and polyvinyl alcohol, or combinations thereof. Yet more preferably, the polymer or copolymer is selected from the group consisting of: polyvinyl alcohol and polyethylene oxide graft copolymers.
Further, in this embodiment, the ratio of acetaminophen to the rate controlling polymer or copolymer or combination thereof is about 1:1 to about 10:1. More preferably, the ratio of acetaminophen to the rate controlling polymer or copolymer or combination thereof is about 3:1 to about 5:1. As provided in the present invention, in one preferred embodiment, the non-core layer has at least one of the following characteristics:
(a) substantially does not crack after 3 months at 40 C, 75% relative humidity in induction-sealed HDPE bottles;
(b) substantially dry (not sticky);
provides fast dissolution in 0.01N HC1 at 37 C to expose the core layer releases at least 80% of the acetaminophen in the non-core layer within 20 minutes of administration to a human patient; or (e) provides a white pigmentation to the formulation without additional pigments.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone of about 0.18 ng/mL/mg to about 1.51 ng/mL/mg, and a plasma concentration at 1 hour Cl for acetaminophen of about 2.34 ng/mL/mg to about 7.24 ng/mL/mg.

In preferred embodiments such as Formulation 15, the dosage form produces a C
l for hydrocodone of about 0.32 ng/mL/mg to about 1.51 ng/mL/mg and a Cl for acetaminophen of about 2.34 ng/mL/mg to about 5.50 ng/mL/mg.
In certain other embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting.
Preferably when administered to a human patient the pharmaceutical composition produces a plasma concentration at 1 hour (Cl) for hydrocodone from about 0.30 ng/mL/mg to about 1.06 ng/mL/mg, and a C l for acetaminophen from about 2.75 ng/mL/mg to about 5.57 ng/mL/mg. In preferred embodiments, the dosage from produces a C l for hydrocodone from about 0.45 ng/mL/mg to about 1.06 ng/mL/mg and a C l for acetaminophen from about 2.75 ng/mL/mg to about 4.43 ng/mL/mg.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.18 g/mL to about 3.63 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined C l for hydrocodone and acetaminophen from about 1.18 g/mL to about 2.76 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In certain embodiments, the dosage form produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.79 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. In preferred embodiments, the dosage from produces a combined Cl for hydrocodone and acetaminophen from about 1.38 g/mL to about 2.23 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen.
In preferred embodiments, the dosage form produces a combined C l for hydrocodone and acetaminophen of 1.80 0.42 g/mL with the 95% confidence interval for the mean value falling between about 1.61 g/mL to about 2.00 g/mL, after a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen. The 95%
confidence interval of combined Cl for hydrocodone and acetaminophen for the preferred embodiments and the Control overlapped. The 95% confidence interval for the mean value of combined Cl for hydrocodone and acetaminophen for the Control ranged from about 1.46 to 1.96 g/mL, after administered as a single dose of 15 mg hydrocodone bitartrate pentahemihydrate and 500 mg of acetaminophen to the human patient. The Control provides sufficient plasma levels of opioid and nonopioid analgesic to provide a reduction in pain intensity within about 1 hour after administration.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. Preferably when administered to a human patient the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose. In another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose. In yet another embodiment, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6 ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose.
In certain embodiments, the following pharmacokinetic profile is preferably exhibited when the single dose comprises about 15 mg of hydrocodone bitartrate pentahemihydrate and about 500 mg of acetaminophen, administered to the patient, when fasting. When administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg. In another embodiment, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg. In yet another embodiment, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
Preferably in this embodiment, the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone.
When administered to a population of healthy North Americans or Western Europeans, particularly when the formulation is adapted to be suitable for, or intended for, administration to a human every 12 hours as needed, about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C. In another embodiment, about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about lhour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about lhour in 0.01 N HC1 at 50 rpm at 37 C.
Further, in another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60%
to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours. In another embodiment, at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and at least 90%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours. In another embodiment, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and at least 95%
of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours. Yet in another embodiment, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours. In another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about l lhours to about 12 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours. In yet another embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
However, when the a slow-release version of the formulation is adapted to be suitable for, or intended for administration to a human, twice daily, as needed, then at least 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours. In another embodiment of the slow release formulation, at least 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours. In another embodiment of the slow release formulation, at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours. In another embodiment of this slow release embodiment, at least 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours. In yet another embodiment of the slow release formulation, at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
In a preferred embodiment, the present invention provides a composition where the core layer comprises an excipient capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug. Further, in a preferred embodiment, the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer. Most preferably, the composition comprises about 500mg of acetaminophen and about 15 mg of hydrocodone bitartrate pentahemihydrate.
In a preferred embodiment, the composition is characterized by at least one of the following features:
i) the amount of abuse-relevant drug that is extracted from the composition by 40% aqueous ethanol within one hour at 37 C in vitro is less than or equal 1.5 times the amount of the hydrocodone that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the hydrocodone and not more than 45% of the hydrocodone during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the acetaminophen within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the acetaminophen and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the hydrocodone upon grinding increases by less than 2- to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37 C , vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0.1 to about 0.2.
The foregoing detailed description and accompanying examples are merely illustrative and not intended as limitations upon the scope of the invention, which is defined solely by the appended claims and their equivalents. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and are part of the present invention. Such changes and modifications, including without limitation those relating to the chemical structures, substituents, derivatives, intermediates, syntheses, formulations and/or methods of use of the invention, can be made without departing from the spirit and scope thereof.

Claims (92)

1. A pharmaceutical composition having a core and a non-core layer, comprising:
(a) hydrocodone, a pharmaceutically acceptable salt or a hydrate thereof, and (b) acetaminophen or ibuprofen, wherein at least 75% all of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core, wherein the acetaminophen or the ibuprofen is the non-core layer, and wherein the composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily.
2. The composition of claim 1, wherein greater than 90% of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core.
3. The composition of claim 1, wherein substantially all of the hydrocodone, pharmaceutically acceptable salt or hydrate thereof is in the core.
4. The composition of claim 1, further wherein the core further comprises acetaminophen.
5. The composition of claim 1, further wherein the core further comprises acetaminophen or ibuprofen.
6. The composition of any one of the claims 1 to 5, wherein when administered to the human patient the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose.
7. The composition of any one of the claims 1 to 5, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose.
8. The composition of any one of the claims 1 to 5, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6 ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose.
9. The composition of any one of the claims 1 to 5, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg.
10. The composition of any one of the claims 1 to 5, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg.
11. The composition of any one of the claims 1 to 5, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
12. The composition of any one of the claims 1 to 5, wherein the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone.
13. The composition of any one of the claims 1 to 5, wherein about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about 1hour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 1 hour in 0.01 NHC1 at 50 rpm at 37°C.
14. The composition of any one of the claims 1 to 5, wherein about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about 1hour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 1 hour in 0.01 NHC1 at 50 rpm at 37°C.
15. The composition of any one of the claims 1 to 5, wherein at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60% to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours.
16. The composition of any one of the claims 1 to 5, wherein at least 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours.
17. The composition of any one of the claims 1 to 5, wherein at least 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and at least 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours.
18. The composition of any one of the claims 1 to 5, wherein at least 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours.
19. The composition of any one of the claims 1 to 5, wherein at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours.
20. The composition of any one of the claims 1 to 5, wherein at least 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours.
21. The composition of any one of the claims 1 to 5, wherein at least 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and at least 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours.
22. The composition of any one of the claims 1 to 5, wherein at least 99% of the hydrocodone is released from the pharmaceutical composition in about 11hours to about 12 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours.
23. The composition of any one of the claims 1 to 5, wherein at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
24. The composition of any one of the claims 1 to 5, wherein at least 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours.
25. The composition of any one of the claims 1 to 5, wherein at least 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and at least 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
26. The composition of any one of claim 1 or 5, wherein the core layer comprises an excipient capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug.
27. The composition of any one of the claims 1 to 5, wherein the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer.
28. The composition of any one of the claims 1 to 27, wherein the composition comprises about 500mg of acetaminophen and about 15 mg of hydrocodone bitartrate pentahemihydrate.
29. A pharmaceutical composition having a core and a non-core layer, comprising:
(a) an abuse-relevant drug, a pharmaceutically acceptable salt or a hydrate thereof and a non-abuse-relevant drug or a pharmaceutically acceptable salt thereof in the core layer, and (b) a non-abuse-relevant drug, a pharmaceutically acceptable salt or a hydrate thereof in the non-core layer, wherein the composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily; and wherein the composition is characterized by at least one of the following features:
i) the amount of abuse-relevant drug that is extracted from the composition by 40% aqueous ethanol within one hour at 37 °C in vitro is less than or equal 1.5 times the amount of the abuse-relevant drug that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 °C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the abuse-relevant drug and not more than 45% of the abuse-relevant drug during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the non-abuse relevant drug within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the non-abuse relevant drug and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the abuse-relevant drug upon grinding increases by less than 2-to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37°C, vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0.1 to about 0.2.
30. The composition of claim 29, wherein the amount of the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 °C is about 70% to about 90% of the amount of the drug that is extracted by 0.01 N
hydrochloric acid within one hour at 37 °C.
31. The composition of claim 29, wherein the amount of the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 °C is about 70% to about 130% of the amount of the drug that is extracted by 0.01 N
hydrochloric acid within one hour at 37 °C.
32. The composition of claim 29, wherein the amount of the abuse-relevant drug that is extracted from the formulation by 40% aqueous ethanol within one hour at 37 °C is about 75% to about 90% of the amount of the drug that is extracted by 0.01 N
hydrochloric acid within one hour at 37 °C.
33. A pharmaceutical composition having a core layer and a non-core layer, (A) wherein the core layer comprises a mixture of:
(a) at least one opioid;
(b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof;
(B) wherein the non-core layer comprises at least one non-opioid analgesic;
and (C) wherein the composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily.
34. The composition of claim 33, wherein the core layer further comprises at least one non-opioid analgesic.
35. The composition of claim 33, wherein the composition is characterized by at least one of the following features:
i) the amount of abuse-relevant drug that is extracted from the composition by 40% aqueous ethanol within one hour at 37 °C in vitro is less than or equal 1.5 times the amount of the abuse-relevant drug that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 °C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the abuse-relevant drug and not more than 45% of the abuse-relevant drug during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the non-abuse relevant drug within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the non-abuse relevant drug and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the abuse-relevant drug upon grinding increases by less than 2-to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37°C , vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0.1 to about 0.2.
36. The composition of claim 33, wherein the opioid is selected from the group consisting of alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levophenacylmorphan, levorphanol, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbulphine, narceine, nicomorphine, norpipanone, opium, oxycodone, oxymorphone, papvretum, pentazocine, phenadoxone, phenazocine, phenomorphan, phenoperidine, piminodine, propiram, propoxyphene, sufentanil, tilidine, and tramadol, and salts, hydrates and mixtures thereof and the non-opioid analgesic is selected from the group consisting of acetaminophen, aspirin, fentaynl, ibuprofen, indomethacin, ketorolac, naproxen, phenacetin, piroxicam, sufentanyl, sunlindac, interferon alpha, and salts, hydrates and mixtures thereof.
37. The composition of claim 33, wherein the opioid is hydrocodone and the non-opioid analgesic is acetaminophen or ibuprofen.
38. The composition of claim 33, wherein the opioid is hydrocodone and the non-opioid analgesic is acetaminophen.
39. The composition of any one of the claims 33 to 38, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose.
40. The composition of any one of the claims 33 to 38, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose.
41. The composition of any one of the claims 33 to 38, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose.
42. The composition of any one of the claims 33 to 38, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg.
43. The composition of any one of the claims 33 to 38, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg.
44. The composition of any one of the claims 33 to 38, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
45. The composition of any one of the claims 33 to 38, wherein the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and zero order or first order for hydrocodone.
46. The composition of any one of the claims 33 to 38, wherein about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about 1hour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 1 hour in 0.01 N HC1 at 50 rpm at 37°C.
47. The composition of any one of the claims 33 to 38, wherein about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about 1hour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 1 hour in 0.01 N HC1 at 50 rpm at 37°C.
48. The composition of any one of the claims 33 to 38, wherein about 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60% to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours.
49. The composition of any one of the claims 33 to 38, wherein about 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and about 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours.
50. The composition of any one of the claims 33 to 38, wherein about 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and about 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours.
51. The composition of any one of the claims 33 to 38, wherein about 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours.
52. The composition of any one of the claims 33 to 38, wherein about 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours.
53. The composition of any one of the claims 33 to 38, wherein about 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours.
54. The composition of any one of the claims 33 to 38, wherein about 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours.
55. The composition of any one of the claims 33 to 38, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in about 11 hours to about 12 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours.
56. The composition of any one of the claims 33 to 38, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
57. The composition of any one of the claims 33 to 38, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours.
58. The composition of any one of the claims 33 to 38, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
59. The composition of any one of the claims 33 to 38, wherein the core layer comprises an excipient capable of controlling the drug release and the non-core layer comprises an excipient capable of instantly releasing the drug.
60. The composition of any one of the claims 33 to 38, wherein the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer.
61. A pharmaceutical composition having a core layer and a non-core layer, (A) wherein the core layer comprises a mixture of (a) at least one opioid and at least one first non-opioid analgesic;
(b) at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof;
(B) wherein the non-core layer comprises at least one second non-opioid analgesic; and (C) wherein the composition is adapted so as to be useful for oral administration to a human 3, 2, or 1 times daily.
62. The composition according to claim 61, wherein the opioid comprises hydrocodone and the first and the second non-opioid analgesic comprises acetaminophen or ibuprofen.
63. The composition according to claim 61, wherein the opioid comprises hydrocodone and the first and the second non-opioid analgesic comprises acetaminophen.
64. The composition according to claim 61, wherein the non-core layer comprises:
acetaminophen; and at least one rate altering pharmaceutically acceptable polymer, copolymer, or a combination thereof.
65. The composition according to claim 64, wherein the polymer or copolymer is selected from the group consisting of: hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose; polymethacrylate, polyvinyl alcohol, polyethylene oxide, and combinations thereof.
66. The composition according to claim 64, wherein the polymer or copolymer is selected from the group consisting of: hydroxypropyl methylcellulose, and polyvinyl alcohol, or combinations thereof.
67. The composition according to claim 64, wherein the polymer or copolymer is selected from the group consisting of: polyvinyl alcohol and polyethylene oxide graft copolymers.
68. The composition according to claim 64, wherein the ratio of acetaminophen to the rate controlling polymer or copolymer or combination thereof is about 1:1 to about 10:1.
69. The composition according to claim 64, wherein the ratio of acetaminophen to the rate controlling polymer or copolymer or combination thereof is about 3:1 to about 5:1.
70. The composition according to any one of the claims 61 to 69, wherein the non-core layer has at least one of the following characteristics:

(a) substantially does not crack after 3 months at 40°C, 75% relative humidity in induction-sealed HDPE bottles;
(b) substantially dry (not sticky);
provides fast dissolution in 0.01N HCl at 37°C to expose the core layer releases at least 80% of the acetaminophen in the non-core layer within 20 minutes of administration to a human patient; or (e) provides a white pigmentation to the formulation without additional pigments.
71. The composition of any one of the claims 61 to 70, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone from about 0.6 ng/mL/mg to about 1.4 ng/mL/mg and a Cmax for acetaminophen from about 2.8 ng/mL/mg and 7.9 ng/mL/mg after a single dose.
72. The composition of any one of the claims 61 to 70, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of about 0.4 ng/mL/mg to about 1.9 ng/mL/mg and a Cmax for acetaminophen of about 2.0 ng/mL/mg to about 10.4 ng/mL/mg after a single dose.
73. The composition of any one of the claims 61 to 70, wherein when administered to the human patient, the pharmaceutical composition produces a plasma profile characterized by a Cmax for hydrocodone of from about 0.6ng/mL/mg to about 1.0 ng/mL/mg and a Cmax for acetaminophen of from about 3.0 ng/mL/mg to about 5.2 ng/mL/mg after a single dose.
74. The composition of any one of the claims 61 to 70, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 9.1 ng*hr/mL/mg to about 19.9 ng*hr/mL/mg and an AUC for acetaminophen of about 28.6 ng*hr/mL/mg to about 59.1 ng*hr/mL/mg.
75. The composition of any one of the claims 61 to 70, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 7.0 ng*hr/mL/mg to about 26.2 ng*hr/mL/mg and an AUC for acetaminophen of about 18.4 ng*hr/mL/mg to about 79.9 ng*hr/mL/mg.
76. The composition of any one of the claims 61 to 70, wherein when administered to the human patient, the dosage form produces an AUC for hydrocodone of about 11.3 ng*hr/mL/mg to about 18.7 ng*hr/mL/mg and an AUC for acetaminophen of about 28.7 ng*hr/mL/mg to about 53.5 ng*hr/mL/mg.
77. The composition of any one of the claims 61 to 70, wherein the in vitro rate of release of the pharmaceutical composition has a biphasic release profile, and wherein for each phase of the in vitro rate of release is zero order or first order for acetaminophen and first order for hydrocodone.
78. The composition of any one of the claims 61 to 70, wherein about 20-45% of the hydrocodone is released in vitro from the pharmaceutical compositions in about 1 hour and about 20-45% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 1 hour in 0.01 N HCl at 50 rpm at 37°C.
79. The composition of any one of the claims 61 to 70, wherein about 25-35% of the hydrocodone is released in vitro from the pharmaceutical compositions in about 1 hour and about 25-35% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 1 hour in 0.01 N HC1 at 50 rpm at 37°C.
80. The composition of any one of the claims 61 to 70, wherein about 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 12 hours and at least 60% to about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 6 hours to about 8.5 hours.
81. The composition of any one of the claims 61 to 70, wherein about 90% of the hydrocodone is released from the pharmaceutical composition in about 18 hours to about 23 hours and about 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 18 hours to about 23 hours.
82. The composition of any one of the claims 61 to 70, wherein about 90% of the hydrocodone is released from the pharmaceutical composition in about 8 hours to about 11 hours and about 90% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 8 hours to about 11 hours.
83. The composition of any one of the claims 61 to 70, wherein about 95% of the hydrocodone is released from the pharmaceutical composition in about 9 hours to about 12 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 9 hours to about 12 hours.
84. The composition of any one of the claims 61 to 70, wherein about 95% is of the hydrocodone is released from the pharmaceutical composition in about 10 hours to about 12 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 10 hours to about 12 hours.
85. The composition of any one of the claims 61 to 70, wherein about 95% of the hydrocodone is released from the pharmaceutical composition in about 20 hours to about 25 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 20 hours to about 25 hours.
86. The composition of any one of the claims 61 to 70, wherein about 95% is of the hydrocodone is released from the pharmaceutical composition in about 21 hours to about 22 hours and about 95% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 21 hours to about 22 hours.
87. The composition of any one of the claims 61 to 70, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in about 11 hours to about 12 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 11 hours to about 12 hours.
88. The composition of any one of the claims 61 to 70, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in less than about 13 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 13 hours.
89. The composition of any one of the claims 61 to 70, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in about 22 hours to about 26 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in about 22 hours to about 26 hours.
90. The composition of any one of the claims 61 to 70, wherein about 99% of the hydrocodone is released from the pharmaceutical composition in less than about 27 hours and about 99% of the acetaminophen is released in vitro from the pharmaceutical compositions in less than about 27 hours.
91. The composition of any one of the claims 61 to 70, wherein the composition is characterized by at least one of the following features:
i) the amount of hydrocodone that is extracted from the composition by 40%
aqueous ethanol within one hour at 37 °C in vitro is less than or equal 1.5 times the amount of the hydrocodone that is extracted by 0.01 N hydrochloric acid in vitro within one hour at 37 °C, ii) the composition does not break under a force of 150 newtons, preferably 300 newtons, more preferably 450 newtons, yet more preferably 500 newtons as measured by "Pharma Test PTB
501" hardness tester, iii) the composition releases at least 20% of the hydrocodone and not more than 45% of the hydrocodone during the first hour of in vitro dissolution testing and preferably also during the first hour of in vivo testing, iv) the composition releases a therapeutically effective dose of the acetaminophen within 1 to 2 hours after a single dose, v) the composition releases a therapeutically effective dose of the acetaminophen and/or the abuse-relevant drug at 1 hour and at 12 hours after a single dose, vi) in the composition, release of the hydrocodone upon grinding increases by less than 2- to 3-fold, as compared to an intact tablet, when the composition is ground for 1 minute by a coffee-grinder at 20,000 - 50,000 rpm, in 40% aqueous ethanol for 1 hour at 37°C , vii) the composition when ground comprises a particulate size of about 2 cm to about 355 micrometer for about 20% of the fraction, greater than about 63 microns and less than about 355 microns for about 66% of the fraction and less than about 63 microns for about 14% of the fraction, as measured by a sieving test, or viii) the composition is substantially smooth, wherein the Centre Line Average (CLA) is from about 0.1 to about 0.6, preferably from about 0.1 to about 0.4, and most preferably from about 0.1 to about 0.2.
92. The composition of any one of the claims 61 to 70, wherein the core layer is manufactured by melt-extrusion followed by direct shaping of the drug-containing melt and the non-core layer is spray coated over the core layer.
CA2690829A 2007-07-20 2007-07-20 Formulations of nonopioid and confined opioid analgesics Abandoned CA2690829A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2007/073957 WO2009014534A1 (en) 2007-07-20 2007-07-20 Formulations of nonopioid and confined opioid analgesics

Publications (1)

Publication Number Publication Date
CA2690829A1 true CA2690829A1 (en) 2009-01-29

Family

ID=39338644

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2690829A Abandoned CA2690829A1 (en) 2007-07-20 2007-07-20 Formulations of nonopioid and confined opioid analgesics

Country Status (14)

Country Link
EP (1) EP2182928A1 (en)
JP (1) JP2010534204A (en)
KR (1) KR20100055431A (en)
CN (1) CN101917977B (en)
AU (1) AU2007356880A1 (en)
BR (1) BRPI0721940A2 (en)
CA (1) CA2690829A1 (en)
IL (1) IL202680A0 (en)
MX (1) MX2010000803A (en)
NZ (1) NZ581767A (en)
RU (1) RU2477995C2 (en)
TW (1) TW200904431A (en)
WO (1) WO2009014534A1 (en)
ZA (1) ZA200908900B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7776314B2 (en) 2002-06-17 2010-08-17 Grunenthal Gmbh Abuse-proofed dosage system
DE10361596A1 (en) 2003-12-24 2005-09-29 Grünenthal GmbH Process for producing an anti-abuse dosage form
DE10336400A1 (en) 2003-08-06 2005-03-24 Grünenthal GmbH Anti-abuse dosage form
DE102005005446A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Break-resistant dosage forms with sustained release
US20070048228A1 (en) 2003-08-06 2007-03-01 Elisabeth Arkenau-Maric Abuse-proofed dosage form
DE102004032049A1 (en) 2004-07-01 2006-01-19 Grünenthal GmbH Anti-abuse, oral dosage form
DE102005005449A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Process for producing an anti-abuse dosage form
DE102007011485A1 (en) 2007-03-07 2008-09-11 Grünenthal GmbH Dosage form with more difficult abuse
RU2493830C2 (en) 2008-01-25 2013-09-27 Грюненталь Гмбх Drug form
AU2009243681B2 (en) 2008-05-09 2013-12-19 Grunenthal Gmbh Process for the preparation of an intermediate powder formulation and a final solid dosage form under usage of a spray congealing step
EP2997965B1 (en) 2009-07-22 2019-01-02 Grünenthal GmbH Tamper-resistant dosage form for oxidation-sensitive opioids
PL2456427T3 (en) 2009-07-22 2015-07-31 Gruenenthal Gmbh Hot-melt extruded controlled release dosage form
TWI516286B (en) 2010-09-02 2016-01-11 歌林達股份有限公司 Tamper resistant dosage form comprising an anionic polymer
MX2013002377A (en) 2010-09-02 2013-04-29 Gruenenthal Gmbh Tamper resistant dosage form comprising inorganic salt.
SG191288A1 (en) * 2010-12-22 2013-07-31 Purdue Pharma Lp Encased tamper resistant controlled release dosage forms
BR112014001091A2 (en) 2011-07-29 2017-02-14 Gruenenthal Gmbh tamper resistant tablet that provides immediate release of the drug
LT2736495T (en) 2011-07-29 2017-11-10 Grünenthal GmbH Tamper-resistant tablet providing immediate drug release
FR2979242A1 (en) * 2011-08-29 2013-03-01 Sanofi Sa COMPRESSES AGAINST ABUSIVE USE, BASED ON PARACETAMOL AND OXYCODONE
BR112014019988A8 (en) 2012-02-28 2017-07-11 Gruenenthal Gmbh BREAK-RESISTANT DOSAGE FORM COMPRISING A PHARMACOLOGICALLY ACTIVE COMPOUND AND AN ANIONIC POLYMER
AR090695A1 (en) 2012-04-18 2014-12-03 Gruenenthal Gmbh PHARMACEUTICAL DOSAGE FORM RESISTANT TO ADULTERATION AND RESISTANT TO IMMEDIATE RELEASE OF DOSE
US10064945B2 (en) 2012-05-11 2018-09-04 Gruenenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
JP6466417B2 (en) * 2013-05-29 2019-02-06 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング A tamper-resistant dosage form with a bimodal release profile
CA2907950A1 (en) 2013-05-29 2014-12-04 Grunenthal Gmbh Tamper-resistant dosage form containing one or more particles
MX368846B (en) 2013-07-12 2019-10-18 Gruenenthal Gmbh Tamper-resistant dosage form containing ethylene-vinyl acetate polymer.
DK3069733T3 (en) * 2013-11-13 2022-11-14 National Defense Education And Res Foundation NEW ACETAMINOPHYDRATE COMPOUND WITH NO LIVER SIDE EFFECTS
CA2931553C (en) 2013-11-26 2022-01-18 Grunenthal Gmbh Preparation of a powdery pharmaceutical composition by means of cryo-milling
CA2947786A1 (en) 2014-05-12 2015-11-19 Grunenthal Gmbh Tamper resistant immediate release capsule formulation comprising tapentadol
EP3148512A1 (en) 2014-05-26 2017-04-05 Grünenthal GmbH Multiparticles safeguarded against ethanolic dose-dumping
EP3285745A1 (en) 2015-04-24 2018-02-28 Grünenthal GmbH Tamper-resistant dosage form with immediate release and resistance against solvent extraction
JP2018520165A (en) * 2015-07-10 2018-07-26 サン、ファーマ、アドバンスト、リサーチ、カンパニー、リミテッドSun Pharma Advanced Research Company Limited Hydrocodone Multiple Tablet Abuse Resistant Immediate Release Solid Dosage Form
JP2018526414A (en) 2015-09-10 2018-09-13 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Protection against oral overdose with abuse-inhibiting immediate release formulations
SE541800C2 (en) 2018-04-20 2019-12-17 Scania Cv Ab A method of controlling a powertrain, a control arrangement, a vehicle, a computer program and a computer-readable medium
JP6858729B2 (en) * 2018-05-25 2021-04-14 ▲財▼▲団▼法人国防教育研究基金会National Defense Education And Research Foundation New acetaminophen complex composition with no side effects on the liver

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU230828B1 (en) * 1999-10-29 2018-08-28 Euro Celtique Sa Controlled release hydrocodone formulations
US20030092724A1 (en) * 2001-09-18 2003-05-15 Huaihung Kao Combination sustained release-immediate release oral dosage forms with an opioid analgesic and a non-opioid analgesic
WO2005030181A1 (en) * 2003-09-26 2005-04-07 Alza Corporation Controlled release formulations of opioid and nonopioid analgesics
NZ546148A (en) * 2003-09-26 2009-05-31 Alza Corp Drug coating providing high drug loading and methods for providing the same
CA2644435C (en) * 2006-03-06 2015-04-07 Pozen Inc. Dosage forms for administering combinations of drugs
US8765178B2 (en) * 2006-07-19 2014-07-01 Watson Laboratories, Inc. Controlled release formulations and associated methods

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10639281B2 (en) 2013-08-12 2020-05-05 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

Also Published As

Publication number Publication date
ZA200908900B (en) 2014-05-28
CN101917977B (en) 2013-05-29
MX2010000803A (en) 2010-06-23
BRPI0721940A2 (en) 2014-03-18
TW200904431A (en) 2009-02-01
KR20100055431A (en) 2010-05-26
IL202680A0 (en) 2010-06-30
JP2010534204A (en) 2010-11-04
NZ581767A (en) 2012-05-25
RU2010106202A (en) 2011-08-27
EP2182928A1 (en) 2010-05-12
RU2477995C2 (en) 2013-03-27
WO2009014534A1 (en) 2009-01-29
AU2007356880A1 (en) 2009-01-29
CN101917977A (en) 2010-12-15

Similar Documents

Publication Publication Date Title
US20170014348A1 (en) Formulations of Nonopioid and Confined Opioid Analgesics
CA2690829A1 (en) Formulations of nonopioid and confined opioid analgesics
US20140120061A1 (en) Abuse resistant melt extruded formulation having reduced alcohol interaction
US20100172989A1 (en) Abuse resistant melt extruded formulation having reduced alcohol interaction
AU2007205866B2 (en) Dosage form and method for the delivery of drugs of abuse
MXPA02004293A (en) Controlled release hydrocodone formulations.
AU2009207579A1 (en) Abuse resistant melt extruded formulation having reduced alcohol interaction
AU2014279743A1 (en) Modified release formulation
SG178771A1 (en) Formulations of nonopioid and confined opioid analgesics

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued

Effective date: 20140722