Methods of manufacturing oral dosage forms

Information

  • Patent Grant
  • 10076516
  • Patent Number
    10,076,516
  • Date Filed
    Tuesday, December 13, 2016
    7 years ago
  • Date Issued
    Tuesday, September 18, 2018
    5 years ago
Abstract
A solid oral controlled-release oral dosage form of hydrocodone is disclosed. The dosage form comprising an analgesically effective amount of hydrocodone or a pharmaceutically acceptable salt thereof, and a sufficient amount of a controlled release material to render the dosage form suitable for twice-a-day administration to a human patient, the dosage form providing a C12/Cmax ratio of 0.55 to 0.85, said dosage form providing a therapeutic effect for at least about 12 hours.
Description
BACKGROUND OF THE INVENTION

Due to the difficulties presented by the pharmacotherapy of pain, particularly chronic pain, opioid analgesics are ideal drugs to be administered as controlled release formulations. The present invention relates to a solid, controlled-release oral dosage form for use in the treatment of pain.


It is the intent of all controlled (slow) release formulations to provide a longer period of pharmacological action after administration than is ordinarily obtained after administration of immediate-release dosage forms. Such longer periods of response provide for many therapeutic benefits that are not achieved with corresponding short acting, immediate release preparations. Thus, therapy may be continued without interrupting the sleep of the patient, which is of special importance, for example, when treating a patient for moderate to severe pain (e.g., a post-surgery patient, a cancer patient, etc.), or for those patients who experience migraine headaches on awakening, as well as for the debilitated patient for whom sleep is essential.


Unless conventional rapid acting drug therapy is carefully administered at frequent intervals to maintain effective steady state plasma concentrations of the drug, peaks and valleys in the plasma level of the active drug occurs because of the rapid absorption, systemic excretion of the compound and through metabolic inactivation, thereby producing special problems in maintenance therapy of the patient. A further general advantage of longer acting drug preparations is improved patient compliance resulting from the avoidance of missed doses through patient forgetfulness.


It is known in the pharmaceutical art to prepare compositions which provide for controlled release of pharmacologically active substances contained in the compositions after oral administration to humans and animals. Such slow release compositions are used to delay absorption of a medicament until it has reached certain portions of the alimentary tract. Such controlled release of a medicament in the alimentary tract further maintains a desired concentration of said medicament in the blood stream for a longer duration than would occur if conventional rapid release dosage forms are administered.


The prior art teaching of the preparation and use of compositions providing the controlled release of an active compound from a carrier is basically concerned with the release of the active substance into the physiologic fluid of the alimentary tract. However, it is generally recognized that the mere presence of an active substance in the gastrointestinal fluids does not, by itself, ensure bioavailability.


In order to be absorbed, the active drug substance must be in solution. The time required for a given proportion of an active substance from a unit dosage form is determined as the proportion of the amount of active drug substance released from a unit dosage form over a specified time base by a test method conducted under standardized conditions. The physiologic fluids of the gastrointestinal tract are the media for determining dissolution time. The present state of the art recognizes many satisfactory test procedures to measure dissolution time for pharmaceutical compositions, and these test procedures are described in official compendia worldwide.


Although there are many diverse factors which influence the dissolution of a drug substance from its carrier, the dissolution time determined for a pharmacologically active substance from the specific composition is relatively constant and reproducible. Among the different factors which may affect the dissolution time are the surface area of the drug substance presented to the dissolution solvent medium, the pH of the solution, the solubility of the substance in the specific solvent medium, and the driving forces of the saturation concentration of dissolved materials in the solvent medium. Thus, the dissolution concentration of an active drug substance is dynamically modified in its steady state as components are removed from the dissolution medium through absorption across the tissue site. Under physiologic conditions, the saturation level of the dissolved materials is replenished from the dosage form reserve to maintain a relatively uniform and constant dissolution concentration in the solvent medium providing for a steady state absorption.


The transport across a tissue absorption site of the gastrointestinal tract is influenced by the Donnan osmotic equilibrium forces on both sides of the membrane since the direction of the driving force is the difference between the concentrations of active substance on either side of the membrane, i.e., the amount dissolved in the gastrointestinal fluids and the amount present in the blood. Since the blood concentrations are constantly being modified by dilution, circulatory changes, tissue storage, metabolic conversion and systemic excretion, the flow of active materials is directed from the gastrointestinal tract into the blood stream.


Various techniques have been used to prepare controlled release dosage forms. Specially coated pellets, tablets and capsules wherein the slow release of the active medicament is brought about through selective breakdown of the coating of the preparation or through compounding with a special matrix to affect the release of a drug are known in the art. Certain controlled release formulations provide for related sequential release of a single dose of an active compound at predetermined periods after administration.


Specific examples of controlled release opioid formulations reported in the patent literature include, for example, those disclosed in U.S. Pat. Nos. 4,990,341 and 4,844,909 (Goldie, et al.), both assigned to the assignee of the present invention and incorporated herein by reference, describe hydromorphone compositions wherein the dissolution rate in-vitro of the dosage form, when measured by the USP Paddle or Basket Method at 100 rpm in 900 ml aqueous buffer (pH between 1.6 and 7.2) at 37° C., is between 12.5 and 42.5% (by wt) hydromorphone released after 1 hour, between 25 and 55% (by wt) released after 2 hours, between 45 and 75% (by wt) released after 4 hours and between 55 and 85% (by wt) released after 6 hours, the in-vitro release rate being independent of pH between pH 1.6 and 7.2 and chosen such that the peak plasma concentration of hydromorphone obtained in-vivo occurs between 2 and 4 hours after administration of the dosage form. At least 12 hours of pain relief is obtained with these hydromorphone formulations.


It is considered highly desirable to provide controlled-release dosage formulations of other opioid analgesic drugs which can be used for moderate pain. It is further considered highly desirable to provide such controlled-release formulations with pharmacokinetic properties which provide the most effective pain management in patients in need of pain therapy.


SUMMARY OF THE INVENTION

It is an object of the present invention to substantially improve the efficiency and quality of pain management in human patients experiencing moderate pain.


It is an object of the present invention to provide bioavailable hydrocodone formulations that substantially improve the efficiency and quality of pain management.


It is yet another object of the present invention to provide bioavailable controlled-release hydrocodone formulations which provide a substantially increased duration of effect as compared to immediate release hydrocodone formulations, but which provide an early onset of analgesia.


It is a further object of the invention to provide orally administrable controlled release opioid formulations suitable for twice-a-day administration which provide an early onset of therapeutic effect and which, after rising to a maximum concentration during the dosage interval, provide a relatively flat serum plasma profile, meaning that the plasma level of the opioid provides a C12/Cmax ratio of 0.55 to 0.85, and which provides effective pain relief to the patient. In alternate embodiments, the dosage form provides a C12/Cmax ratio of 0.65 to 0.75


The above objects and others are attained by virtue of the present invention, which in certain embodiments, provides a solid oral controlled-release dosage form comprising an analgesically effective amount of hydrocodone or a pharmaceutically acceptable salt thereof and a sufficient amount of a controlled release material to render the dosage form suitable for twice-a-day administration, the dosage form after single administration to a human patient or a population of patients, providing a time to peak plasma concentration of hydrocodone in-vivo, preferably at from about 2 to about 8 hours (Tmax), and after attaining a maximum concentration, providing a C12/Cmax ratio of 0.55 to 0.85.


In certain preferred embodiments, the controlled release dosage form provides an in-vitro release of from 18% to about 42.5% by weight of the hydrocodone or salt thereof from the dosage form at one hour when measured by the USP Basket Method at 100 rpm in 700 ml of Simulated Gastric Fluid (SGF) for 55 minutes at 37° C. and thereafter switching to 900 ml of Simulated Intestinal Fluid (SIF) at 37° C.


In certain preferred embodiments, the dissolution rate in-vitro of the hydrocodone dosage form when measured by the USP Basket method at 100 rpm in 900 ml aqueous buffer at a pH of 1.2 and 7.5 at 37° C. is from about 25 to about 65% by weight of the hydrocodone or salt thereof released after 2 hours, from about 45 to about 85% by weight of the hydrocodone or salt thereof released after 4 hours, and greater than about 60% by weight of the hydrocodone or salt thereof released after 8 hours. Although the in-vitro release rate may be either pH-independent or pH-dependent as desired, in preferred embodiments of the invention the release of hydrocodone is pH-independent.


In certain preferred embodiments, there is provided a controlled release dosage form comprising a therapeutically effective amount of hydrocodone wherein the dosage form provides a hydrocodone plasma concentration of at least 5 or 6 ng/ml, at 12 hours after administration and provides a plasma concentration of at least about 8 ng/ml at from about 2 to about 8 hours after administration.


In other preferred embodiments of the invention, there is provided a twice-a-day oral controlled release dosage form of hydrocodone which provides a Cmax of hydrocodone which is less than about 50% of the Cmax of an equivalent dose of an immediate release hydrocodone reference formulation (e.g. Lortab®), and which provides effective analgesia during the 12 hour dosage interval.


In other preferred embodiments of the invention, there is provided a twice-a-day controlled release dosage form of hydrocodone wherein the dosage form provides a time to 80% Cmax which is from about 90% to about 150%, preferably from about 90% to about 110%, of the time to 80% Cmax of an equivalent dose of immediate release hydrocodone reference formulation (e.g. Lortab). Preferably, the time to 80% Cmax of hydrocodone for the controlled release dosage form being from about 0.5 to about 1.5 hours, most preferably from about 0.8 to about 1.2 hours. In alternate embodiments, the time to 80% Cmax of hydrocodone for the controlled release dosage form is from about 0.75 to about 2.0 hours, most preferably from about 0.9 to about 1.5 hours.


In other preferred embodiments of the invention, there is provided a twice-a-day controlled release dosage form of hydrocodone wherein the dosage form provides a time to 90% Cmax which is about 150% to about 400%, preferably from about 150% to about 250%, of the time to 90% Cmax of an equivalent dose of immediate release hydrocodone reference formulation. Preferably, the time to 90% Cmax of hydrocodone for the controlled release dosage form is from about 1.5 to about 2.5 hours, most preferably from about 1.8 to about 2.2 hours. In alternate embodiments, the time to 90% Cmax of hydrocodone for the controlled release dosage form is from about 1.5 to about 4.0 hours, most preferably from about 1.8 to about 2.5 hours.


In other preferred embodiments of the invention, there is provided a twice-a-day controlled release dosage form of hydrocodone wherein the dosage form maintains a plasma concentration within 80% of Cmax from about 0.5 to 10 hours, preferably from about 1 to about 9 hours or from about 4 to about 8 hours.


In other preferred embodiments of the invention, there is provided a twice-a-day controlled release dosage form of hydrocodone which maintains a plasma concentration of hydrocodone within 90% of Cmax from about 1 to 6.5 hours, preferably from about 2 to about 5 hours or from about 2 to about 6.5 hours.


In other preferred embodiments of the invention, there is provided a twice-a-day controlled release dosage form of hydrocodone which provides a mean in-vivo absorption rate from administration to Tmax from about 1.5 mg/hour to about 5 mg/hour and provides a mean rate of absorption from Tmax to the end of the dosing interval which is less than about 0.5 mg/hour based on oral administration of a dosage form containing 15 mg hydrocodone bitartrate. Preferably, the dosage form provides a mean in-vivo absorption rate from administration to Tmax from about 2 mg/hour to about 4 mg/hour and provides a mean in-vivo absorption rate Tmax to the end of the 12 hour dosing interval which is from about 0.08 mg/hour to about 0.4 mg/hour based on oral administration of a dosage form containing 15 mg hydrocodone bitartrate.


In other preferred embodiments of the invention, there is provided a twice-a-day oral controlled release hydrocodone dosage form which provides a rate of absorption during the time period from Tmax to about 12 hours after oral administration of the dosage form which is from about 55% to about 85% of the rate of elimination during the same time period.


The above embodiments of the invention, as well as other embodiments, preferably provide a time to Tmax at a time point 3 to 4 times later than the Tmax provided by an equivalent dose of an immediate release hydrocodone reference. Preferably, the Tmax provided by the sustained release formulation occurs at from about 2 to about 8 hours, from about 3 to about 7 hours or from about 4 to about 6 hours after oral administration.


The present invention is further directed to hydrocodone formulations which provide a Cmax of hydrocodone which is less than about 50%, preferably less than about 40% of the Cmax provided by an equivalent dose of an immediate release reference product.


For example, it was surprisingly discovered that when hydrocodone is formulated in the delivery system as disclosed in U.S. Pat. Nos. 4,861,598 and 4,970,075, the Cmax of hydrocodone provided by the delivery system as a percentage of the Cmax of an immediate release reference product was considerably lower than the same calculation for oxycodone formulated in the same delivery system. This phenomena is evident, regardless of the fact that the controlled release oxycodone and hydrocodone formulations exhibited similar in-vitro dissolution parameters.


When the present invention is formulated using the delivery systems U.S. Pat. Nos. 4,861,598 and 4,970,075, the Cmax of the delivery system as a percentage of the Cmax of the immediate release reference product is less than about 50%, and less than 40% in preferred embodiments, whereas oxycodone, exhibits a calculation of greater than 50%.


“Hydrocodone” is defined for purposes of the invention as including hydrocodone free base, as well as pharmaceutically acceptable salts and complexes of hydrocodone.


The term “USP Paddle or Basket Method” is the Paddle and Basket Method described, e.g., in U.S. Pharmacopoeia XXII (1990), herein incorporated by reference.


The term “pH-dependent” for purposes of the present invention is defined as having characteristics (e.g. dissolution) which vary according to environmental pH.


The term “pH-independent” for purposes of the present invention is defined as having characteristics (e.g., dissolution) which are substantially unaffected by pH.


The term “bioavailability” is defined for purposes of the present invention as the extent to which the drug (e.g., hydrocodone) is absorbed from the unit dosage forms.


The term “controlled-release” is defined for purposes of the present invention as the release of the drug (e.g., hydrocodone) at such a rate that blood (e.g., plasma) concentrations are maintained within the therapeutic range but below toxic concentrations over a period of time of about 12 hours or longer.


The term “Cmax” denotes the maximum plasma concentration obtained during the dosing interval.


The term “Tmax” denotes the time to maximum plasma concentration (Cmax).


The term T1/2 (abs) denotes the amount of time necessary for one-half of the absorbable dose of opioid to be transferred to plasma.


The term “steady state” means that a plasma concentration for a given drug has been achieved and which is maintained with subsequent doses of the drug at a concentration which is at or above the minimum effective therapeutic concentration and is below the minimum toxic plasma concentration for a given drug. For opioid analgesics, the minimum effective therapeutic concentration will be a partially determined by the amount of pain relief achieved in a given patient. It will be well understood by those skilled in the medical art that pain measurement is highly subjective and great individual variations may occur among patients.


The terms “maintenance therapy” and “chronic therapy” are defined for purposes of the present invention as the drug therapy administered to a patient after a patient is titrated with an opioid analgesic to a steady state as defined above.


The term “minimum effective analgesic concentration” or “MEAC” with respect to concentrations of opioids such as hydrocodone is very difficult to quantify. However, there is generally a minimally effective analgesic concentration of plasma hydrocodone below which no analgesia is provided. While there is an indirect relationship between, e.g., plasma hydrocodone levels and analgesia, higher and prolonged plasma levels are generally associated with superior pain relief. There is a lag time or hysteresis, between the time of peak plasma hydrocodone-levels and the time of peak drug effects. This holds true for the treatment of pain with opioid analgesics in general.


The term custom charactermean resonance timecustom character (MRT) is defined as the average time a drug molecule stays in the body. This calculation, which is a function of absorption, distribution and elimination, is dependent in part, on the dosage form containing the active ingredient.


For purposes of the invention, unless further specified, the term “a patient” means that the discussion (or claim) is directed to the pharmacokinetic parameters of an individual patient or subject.


The term “population of patients” means that the discussion (or claim) is directed to the mean pharmacokinetic parameters of at least two patients or subjects.


The term custom characterbreakthrough paincustom character means pain which the patient experiences despite the fact that the patient is being administered generally effective amounts of the sustained release solid oral dosage forms of the invention containing hydromorphone.


The term custom characterrescuecustom character refers to a dose of an analgesic which is administered to a patient experiencing breakthrough pain.


The term custom charactereffective pain managementcustom character means an objective evaluation of a human patient's response (pain experienced versus side effects) to analgesic treatment by a physician as well as subjective evaluation of therapeutic treatment by the patient undergoing such treatment. One skilled in the art will understand that effective analgesia will vary according to many factors, including individual patient variability.


The term “immediate release hydrocodone reference formulation” for purposes of the present invention, is an equivalent amount of the hydrocodone portion of Lortab®, commercially available from UCB Pharma, Inc, or a pharmaceutical product that provides an immediate release of hydrocodone or a salt thereof.


For purposes of the invention, the controlled release formulations disclosed herein and the immediate release control formulations are dose proportional. In such formulations, the pharmacokinetic parameters (e.g. AUC and Cmax) increase linearly from one dosage strength to another. Therefore the pharmacokinetic parameters of a particular dose can be inferred from the parameters of a different dose of the same formulation.


For purposes of the invention, unless otherwise specified, the pharmacokinetic parameters disclosed herein are based on the administration of a single dose of a hydrocodone formulation to an individual patient. Pharmacokinetic parameters based on a patient population will be specified as “mean” data.


The term “first administration” means a single dose of the present invention at the initiation of therapy to an individual patient or a patient population.


The controlled-release oral solid dosage forms of the present invention surprisingly may be opioid-sparing. It is possible that the controlled-release oral solid dosage forms of the present invention may be dosed at a substantially lower daily dosage in comparison to conventional immediate-release products, with no difference in analgesic efficacy. At comparable daily dosages, greater efficacy may result with the use of the controlled-release oral solid dosage forms of the present invention in comparison to conventional immediate-release products.





BRIEF DESCRIPTION OF THE DRAWINGS

The figures attached herewith are illustrative of embodiments of the invention and are not meant to limit the scope of the invention as encompassed by the claims.



FIG. 1 is a graphical representation of the mean hydrocodone plasma concentration of Example 1, Example 2, Example 3 and an equivalent dose of immediate release hydrocodone.



FIG. 2 is a graphical representation of the mean plasma concentration of Example 1, Example 2 and Example 3, against different samples of controlled release oxycodone manufactured in accordance with the procedures of Example 4, and different samples of controlled release morphine manufactured in accordance with the procedures of Example 5.



FIG. 3 is a graphical representation of the % fraction hydrocodone absorbed over time of Example 1, Example 2, Example 3 and an equivalent dose of immediate release hydrocodone.





DETAILED DESCRIPTION

The above embodiments of the invention can be provided by a wide variety of controlled release formulations known to those skilled in the art. For example, suitable controlled release dosage forms are disclosed in U.S. Pat. Nos. 4,861,598 and 4,970,075, hereby incorporated by reference


In certain embodiments of the present invention, an effective amount of opioid in immediate release form is included in the formulation. The immediate release form of the opioid is included in an amount which is effective to shorten the time to maximum concentration of the opioid in the blood (e.g., plasma), such that the Tmax is shortened to a time of, e.g., from about 2 to about 5 hours, or from about 2 to about 4 hours. It has been discovered that by including such an effective amount of immediate release opioid in the unit dose, the experience of relatively higher levels of pain in patients is significantly reduced. In such embodiments, an effective amount of the opioid in immediate release form may be coated onto the substrates of the present invention. For example, where the extended release opioid from the formulation is due to a controlled release coating, the immediate release layer would be overcoated on top of the controlled release coating. On the other hand, the immediate release layer may be coated onto the surface of substrates wherein the opioid is incorporated in a controlled release matrix. Where a plurality of the sustained release substrates comprising an effective unit dose of the opioid (e.g., multiparticulate systems including pellets, spheres, beads and the like) are incorporated into a hard gelatin capsule, the immediate release portion of the opioid dose may be incorporated into the gelatin capsule via inclusion of the sufficient amount of immediate release opioid as a powder or granulate within the capsule. Alternatively, the gelatin capsule itself may be coated with an immediate release layer of the opioid. One skilled in the art would recognize still other alternative manners of incorporating the immediate release opioid portion into the unit dose. Such alternatives are deemed to be encompassed by the appended claims.


One advantage of the opioid dosage forms of the present invention is that therapeutic concentrations are generally achieved substantially without significant increases in the intensity and/or degree of concurrent side effects, such as nausea, vomiting, or drowsiness, which are often associated with high blood concentrations of opioids. There is also evidence to suggest that the use of the present dosage forms lead to a reduced risk of drug addiction.


Active Agent

The controlled release oral dosage forms of the present invention preferably include from about 0.5 mg to about 1250 mg hydrocodone or an equivalent amount of a pharmaceutically acceptable salt thereof. In more preferred embodiments, the dosage form can include from about 5 mg to about 60 mg, e.g. 15 mg. Suitable pharmaceutically acceptable salts of hydrocodone include hydrocodone bitartrate, hydrocodone bitartrate hydrate, hydrocodone hydrochloride, hydrocodone p-toluenesulfonate, hydrocodone phosphate, hydrocodone thiosemicarbazone, hydrocodone sulfate, hydrocodone trifluoroacetate, hydrocodone hemipentahydrate, hydrocodone pentafluoropropionate, hydrocodone p-nitrophenylhydrazone, hydrocodone o-methyloxime, hydrocodone semicarbazone, hydrocodone hydrobromide, hydrocodone mucate, hydrocodone oleate, hydrocodone phosphate dibasic, hydrocodone phosphate monobasic, hydrocodone inorganic salt, hydrocodone organic salt, hydrocodone acetate trihydrate, hydrocodone bis(heptafluorobutyrate), hydrocodone bis(methylcarbamate), hydrocodone bis(pentafluoropropionate), hydrocodone bis(pyridine carboxylate), hydrocodone bis(trifluoroacetate), hydrocodone chlorhydrate, and hydrocodone sulfate pentahydrate. Preferably, the hydrocodone is present as the bitartrate salt.


The dosage forms of the present invention may further include one or more additional drugs which may or may not act synergistically with the hydrocodone analgesics of the present invention. Examples of such additional drugs include non-steroidal anti-inflammatory agents, including ibuprofen, diclofenac, naproxen, benoxaprofen, flurbiprofen, fenoprofen, flubufen, ketoprofen, indoprofen, piroprofen, carprofen, oxaprozin, pramoprofen, muroprofen, trioxaprofen, suprofen, aminoprofen, tiaprofenic acid, fluprofen, bucloxic acid, indomethacin, sulindac, tolmetin, zomepirac, tiopinac, zidometacin, acemetacin, fentiazac, clidanac, oxpinac, mefenamic acid, meclofenamic acid, flufenamic acid, niflumic acid tolfenamic acid, diflurisal, flufenisal, piroxicam, sudoxicam or isoxicam, and the like. Such non-steroidal anti-inflammatory agents also include cyclo-oxygenase inhibitors such as celecoxib (SC-58635), DUP-697, flosulide (CGP-28238), meloxicam, 6-methoxy-2 naphthylacetic acid (6-MNA), Vioxx (MK-966), nabumetone (prodrug for 6-MNA), nimesulide, NS-398, SC-5766, SC-58215, and T-614. as amantadine (1-aminoadamantine), and memantine (3,5 dimethylaminoadamantone), their mixtures and pharmaceutically acceptable salts thereof.


Other additional drugs include nontoxic NMDA receptor antagonists such dextrorphan, dextromethorphan, 3-(1-naphthalenyl)-5-(phosphonomethyl)-L-phenylalanine, 3-(1-naphthalenyl)-5-(phosphonomethyl)-DL-phenylalanine, 1-(3,5-dimethylphenyl)naphthalene, and 2-(3,5-dimethylphenyl) naphthalene, 2SR,4RS-4-(((1H-Tetrazol-5-yl)methyl)oxy)piperidine-2-carboxylic acid; 2SR,4RS-4-((((1H-Tetrazol-5-yl)methyl)oxy)methyl)piperidine-2-carboxylic acid; E and Z 2SR-4-(O-(1H-Tetrazol-5-yl)methyl)ketoximino)piperidine-2-carboxylic acid; 2SR,4RS-4-((1H-Tetrazol-5-yl)thio)piperidine-2-carboxylic acid; 2SR,4RS-4-((1H-Tetrazol-5-yl)thio)piperidine-2-carboxylic acid; 2SR,4RS-4-(5-mercapto-1H-Tetrazol-1-yl)piperidine-2-carboxylic acid; 2SR,4RS-4-(5-mercapto-2H-Tetrazol-2-yl)piperidine-2-carboxylic acid; 2SR,4RS-4-(5-mercapto-1H-Tetrazol-1-yl)piperidine-2-carboxylic acid; 2SR,4RS-4-(5-mercapto-2H-Tetrazol-2-yl)piperidine-2-carboxylic acid; 2SR,4RS-4-(((1H-Tetrazol-5-yl)thio)methyl)piperidine-2-carboxylic acid; 2SR,4RS-4-((5-mercapto-1H-Tetrazol-1-yl)methyl)piperidine-2-carboxylic acid; or 2SR,4RS-4-((5-mercapto-2H-Tetrazol-2-yl)methyl)piperidine-2-carboxylic acid, their mixtures and pharmaceutically acceptable salts thereof.


Other suitable additional drugs which may be included in the dosage forms of the present invention include acetaminophen, aspirin, neuro-active steroids (such as those disclosed in U.S. Ser. No. 09/026,520, filed Feb. 20, 1998, hereby incorporated by reference) and other non-opioid analgesics.


For example, if a second (non-opioid) drug is included in the formulation, such drug may be included in controlled release form or in immediate release form. The additional drug may be incorporated into the controlled release matrix along with the opioid; incorporated into the controlled release coating; incorporated as a separated controlled release layer or immediate release layer; or may be incorporated as a powder, granulation, etc., in a gelatin capsule with the substrates of the present invention.


In certain preferred embodiments of the present invention, an effective amount of hydrocodone in immediate release form is included in the controlled release unit dose hydrocodone formulation to be administered. The immediate release form of the hydrocodone is included in an amount which is effective to shorten the time to Cmax of the hydrocodone in the blood (e.g., plasma). In such embodiments, an effective amount of the hydrocodone in immediate release form may be coated onto the substrates of the present invention. For example, where the extended release hydrocodone from the formulation is due to a controlled release coating, the immediate release layer would be overcoated on top of the controlled release coating. On the other hand, the immediate release layer may be coated onto the surface of substrates wherein the hydrocodone is incorporated in a controlled release matrix. Where a plurality of the sustained release substrates comprising an effective unit dose of the hydrocodone (e.g., multiparticulate systems including pellets, spheres, beads and the like) are incorporated into a hard gelatin capsule, the immediate release portion of the opioid dose may be incorporated into the gelatin capsule via inclusion of the sufficient amount of immediate release hydrocodone as a powder or granulate within the capsule. Alternatively, the gelatin capsule itself may be coated with an immediate release layer of the hydrocodone. One skilled in the art would recognize still other alternative manners of incorporating the immediate release hydromorphone portion into the unit dose. Such alternatives are deemed to be encompassed by the appended claims. It has been discovered that by including such an effective amount of immediate release hydrocodone in the unit dose, the experience of relatively higher levels of pain in patients is significantly reduced.


Dosage Forms

The controlled-release dosage form may optionally include a controlled release material which is incorporated into a matrix along with the hydrocodone, or which is applied as a sustained release coating over a substrate comprising the drug (the term “substrate” encompassing beads, pellets, spheroids, tablets, tablet cores, etc). The controlled release material may be hydrophobic or hydrophilic as desired. The oral dosage form according to the invention may be provided as, for example, granules, spheroids, pellets (hereinafter collectively referred to as “multiparticulates”). An amount of the multiparticulates which is effective to provide the desired dose of opioid over time may be placed in a capsule or may be incorporated in any other suitable oral solid form, e.g., compressed into a tablet. On the other hand, the oral dosage form according to the present invention may be prepared as a tablet core coated with a controlled-release coating, or as a tablet comprising a matrix of drug, controlled release material, and optionally other pharmaceutically desirable ingredients (e.g., diluents, binders, colorants, lubricants, etc.).


Controlled Release Matrix Formulations

In certain preferred embodiments of the present invention, the controlled-release formulation is achieved via a matrix (e.g. a matrix tablet) which includes a controlled-release material as set forth above. A dosage form including a controlled-release matrix provides in-vitro dissolution rates of the opioid within the preferred ranges and that releases the opioid in a pH-dependent or pH-independent manner. The materials suitable for inclusion in a controlled-release matrix will depend on the method used to form the matrix. The oral dosage form may contain between 1% and 80% (by weight) of at least one hydrophilic or hydrophobic controlled release material.


A non-limiting list of suitable controlled-release materials which may be included in a controlled-release matrix according to the invention include hydrophilic and/or hydrophobic materials, such as gums, cellulose ethers, acrylic resins, protein derived materials, waxes, shellac, and oils such as hydrogenated castor oil, hydrogenated vegetable oil. However, any pharmaceutically acceptable hydrophobic or hydrophilic controlled-release material which is capable of imparting controlled-release of the opioid may be used in accordance with the present invention. Preferred controlled-release polymers include alkylcelluloses such as ethylcellulose, acrylic and methacrylic acid polymers and copolymers, and cellulose ethers, especially hydroxyalkylcelluloses (especially hydroxypropylmethylcellulose) and carboxyalkylcelluloses. Preferred acrylic and methacrylic acid polymers and copolymers include methyl methacrylate, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamine copolymer, poly(methyl methacrylate), poly(methacrylic acid)(anhydride), polymethacrylate, polyacrylamide, poly(methacrylic acid anhydride), and glycidyl methacrylate copolymers. Certain preferred embodiments utilize mixtures of any of the foregoing controlled-release materials in the matrices of the invention.


The matrix also may include a binder. In such embodiments, the binder preferably contributes to the controlled-release of the hydrocodone from the controlled-release matrix.


Preferred hydrophobic binder materials are water-insoluble with more or less pronounced hydrophilic and/or hydrophobic trends. Preferably, the hydrophobic binder materials useful in the invention have a melting point from about 30 to about 200° C., preferably from about 45 to about 90° C. When the hydrophobic material is a hydrocarbon, the hydrocarbon preferably has a melting point of between 25° and 90° C. Of the long chain (C8-C50) hydrocarbon materials, fatty (aliphatic) alcohols are preferred. The oral dosage form may contain up to 80% (by weight) of at least one digestible, long chain hydrocarbon.


Preferably, the oral dosage form contains up to 80% (by weight) of at least one polyalkylene glycol. Specifically, the hydrophobic binder material may comprise natural or synthetic waxes, fatty alcohols (such as lauryl, myristyl, stearyl, cetyl or preferably cetostearyl alcohol), fatty acids, including but not limited to fatty acid esters, fatty acid glycerides (mono-, di-, and tri-glycerides), hydrogenated fats, hydrocarbons, normal waxes, stearic aid, stearyl alcohol and hydrophobic and hydrophilic materials having hydrocarbon backbones. Suitable waxes include, for example, beeswax, glycowax, castor wax and carnauba wax. For purposes of the present invention, a wax-like substance is defined as any material which is normally solid at room temperature and has a melting point of from about 30 to about 100° C.


Preferred hydrophobic binder materials which may be used in accordance with the present invention include digestible, long chain (C8-C50, especially C12-C40), substituted or unsubstituted hydrocarbons, such as fatty acids, fatty alcohols, glyceryl esters of fatty acids, mineral and vegetable oils, natural and synthetic waxes and polyalkylene glycols. Hydrocarbons having a melting point of between 25° and 90° C. are preferred. Of the long-chain hydrocarbon binder materials, fatty (aliphatic) alcohols are preferred in certain embodiments. The oral dosage form may contain up to 80% (by weight) of at least one digestible, long chain hydrocarbon.


In certain preferred embodiments, a combination of two or more hydrophobic binder materials are included in the matrix formulations. If an additional hydrophobic binder material is included, it is preferably selected from natural and synthetic waxes, fatty acids, fatty alcohols, and mixtures of the same. Examples include beeswax, carnauba wax, stearic acid and stearyl alcohol. This list is not meant to be exclusive.


One particular suitable controlled-release matrix comprises at least one water soluble hydroxyalkyl cellulose, at least one C12-C36, preferably C14-C22, aliphatic alcohol and, optionally, at least one polyalkylene glycol. The hydroxyalkyl cellulose is preferably a hydroxy (C1 to C6) alkyl cellulose, such as hydroxypropylcellulose, hydroxypropylmethylcellulose and, especially, hydroxyethyl cellulose. The amount of the at least one hydroxyalkyl cellulose in the present oral dosage form will be determined, inter alia, by the precise rate of opioid release required. The aliphatic alcohol may be, for example, lauryl alcohol, myristyl alcohol or stearyl alcohol. In particularly preferred embodiments of the present oral dosage form, however, the at least one aliphatic alcohol is cetyl alcohol or cetostearyl alcohol. The amount of the aliphatic alcohol in the present oral dosage form will be determined, as above, by the precise rate of opioid release required. It will also depend on whether at least one polyalkylene glycol is present in or absent from the oral dosage form. In the absence of at least one polyalkylene glycol, the oral dosage form preferably contains between 20% and 50% (by wt) of the aliphatic alcohol. When a polyalkylene glycol is present in the oral dosage form, then the combined weight of the aliphatic alcohol and the polyalkylene glycol preferably constitutes between 20% and 50% (by wt) of the total dosage.


In one preferred embodiment, the ratio of, e.g., the at least one hydroxyalkyl cellulose or acrylic resin to the at least one aliphatic alcohol/polyalkylene glycol determines, to a consider-able extent, the release rate of the opioid from the formulation. A ratio of the hydroxyalkyl cellulose to the aliphatic alcohol/polyalkylene glycol of between 1:2 and 1:4 is preferred, with a ratio of between 1:3 and 1:4 being particularly preferred.


The polyalkylene glycol may be, for example, polypropylene glycol or, which is preferred, polyethylene glycol. The number average molecular weight of the at least one polyalkylene glycol is preferred between 1,000 and 15,000 especially between 1,500 and 12,000.


Another suitable controlled-release matrix comprises an alkylcellulose (especially ethylcellulose), a C12 to C36 aliphatic alcohol and, optionally, a polyalkylene glycol.


In addition to the above ingredients, a controlled-release matrix may also contain suitable quantities of other materials, e.g., diluents, lubricants, binders, granulating aids, colorants, flavorants and glidants that are conventional in the pharmaceutical art.


In order to facilitate the preparation of a solid, controlled-release oral dosage form according to this invention there is provided, in a further aspect of the present invention, a process for the preparation of a solid, controlled-release oral dosage form according to the present invention comprising incorporating opioids or a salt thereof in a controlled-release matrix. Incorporation in the matrix may be effected, for example, by


(a) forming granules comprising at least one hydrophobic and/or hydrophilic material as set forth above (e.g., a water soluble hydroxyalkyl cellulose) together with the hydrocodone;


(b) mixing the at least one hydrophobic and/or hydrophilic material-containing granules with at least one C12-C36 aliphatic alcohol, and


(c) optionally, compressing and shaping the granules.


The granules may be formed by any of the procedures well-known to those skilled in the art of pharmaceutical formulation. For example, in one preferred method, the granules may be formed by wet granulating hydroxyalkyl cellulose/opioid with water. In a particularly preferred embodiment of this process, the amount of water added during the wet granulation step is preferably between 1.5 and 5 times, especially between 1.75 and 3.5 times, the dry weight of the opioid.


The matrices of the present invention may also be prepared via a melt pellitization technique. In such circumstance, the opioid in finely divided form is combined with a binder (also in particulate form) and other optional inert ingredients, and thereafter the mixture is pelletized, e.g., by mechanically working the mixture in a high shear mixer to form the pellets (granules, spheres). Thereafter, the pellets (granules, spheres) may be sieved in order to obtain pellets of the requisite size. The binder material is preferably in particulate form and has a melting point above about 40° C. Suitable binder substances include, for example, hydrogenated castor oil, hydrogenated vegetable oil, other hydrogenated fats, fatty alcohols, fatty acid esters, fatty acid glycerides, and the like.


Controlled-release matrices can also be prepared by, e.g., melt-granulation or melt-extrusion techniques. Generally, melt-granulation techniques involve melting a normally solid hydrophobic binder material, e.g. a wax, and incorporating a powdered drug therein. To obtain a controlled release dosage form, it may be necessary to incorporate a hydrophobic controlled release material, e.g. ethylcellulose or a water-insoluble acrylic polymer, into the molten wax hydrophobic binder material. Examples of controlled-release formulations prepared via melt-granulation techniques are found, e.g., in U.S. Pat. No. 4,861,598, assigned to the Assignee of the present invention and hereby incorporated by reference in its entirety.


The additional hydrophobic binder material may comprise one or more water-insoluble wax-like thermoplastic substances possibly mixed with one or more wax-like thermoplastic substances being less hydrophobic than said one or more water-insoluble wax-like substances. In order to achieve controlled release, the individual wax-like substances in the formulation should be substantially non-degradable and insoluble in gastrointestinal fluids during the initial release phases. Useful water-insoluble wax-like binder substances may be those with a water-solubility that is lower than about 1:5,000 (w/w).


In addition to the above ingredients, a controlled release matrix may also contain suitable quantities of other materials, e.g., diluents, lubricants, binders, granulating aids, colorants, flavorants and glidants that are conventional in the pharmaceutical art in amounts up to about 50% by weight of the particulate if desired. The quantities of these additional materials will be sufficient to provide the desired effect to the desired formulation.


Specific examples of pharmaceutically acceptable carriers and excipients that may be used to formulate oral dosage forms are described in the Handbook of Pharmaceutical Excipients, American Pharmaceutical Association (1986), incorporated by reference herein.


The preparation of a suitable melt-extruded matrix according to the present invention may, for example, include the steps of blending the opioid analgesic, together with a controlled release material and preferably a binder material to obtain a homogeneous mixture. The homogeneous mixture is then heated to a temperature sufficient to at least soften the mixture sufficiently to extrude the same. The resulting homogeneous mixture is then extruded, e.g., using a twin-screw extruder, to form strands. The extrudate is preferably cooled and cut into multiparticulates by any means known in the art. The strands are cooled and cut into multiparticulates. The multiparticulates are then divided into unit doses. The extrudate preferably has a diameter of from about 0.1 to about 5 mm and provides controlled release of the therapeutically active agent for a time period of from about 8 to about 24 hours.


An optional process for preparing the melt extrusioned formulations of the present invention includes directly metering into an extruder a hydrophobic controlled release material, a therapeutically active agent, and an optional binder material; heating the homogenous mixture; extruding the homogenous mixture to thereby form strands; cooling the strands containing the homogeneous mixture; cutting the strands into particles having a size from about 0.1 mm to about 12 mm; and dividing said particles into unit doses. In this aspect of the invention, a relatively continuous manufacturing procedure is realized.


Plasticizers, such as those described hereinabove, may be included in melt-extruded matrices. The plasticizer is preferably included as from about 0.1 to about 30% by weight of the matrix. Other pharmaceutical excipients, e.g., talc, mono or poly saccharides, colorants, flavorants, lubricants and the like may be included in the controlled release matrices of the present invention as desired. The amounts included will depend upon the desired characteristic to be achieved.


The diameter of the extruder aperture or exit port can be adjusted to vary the thickness of the extruded strands. Furthermore, the exit part of the extruder need not be round; it can be oblong, rectangular, etc. The exiting strands can be reduced to particles using a hot wire cutter, guillotine, etc. A melt extruded multiparticulate system can be, for example, in the form of granules, spheroids or pellets depending upon the extruder exit orifice. For purposes of the present invention, the terms “melt-extruded multiparticulate(s)” and “melt-extruded multiparticulate system(s)” and “melt-extruded particles” shall refer to a plurality of units, preferably within a range of similar size and/or shape and containing one or more active agents and one or more excipients, preferably including a hydrophobic controlled release material as described herein. Preferably the melt-extruded multiparticulates will be of a range of from about 0.1 to about 12 mm in length and have a diameter of from about 0.1 to about 5 mm. In addition, it is to be understood that the melt-extruded multiparticulates can be any geometrical shape within this size range, such as, simply by way of example, beads, seeds, pellets, etc. Alternatively, the extrudate may simply be cut into desired lengths and divided into unit doses of the therapeutically active agent without the need of a spheronization step.


In one preferred embodiment, oral dosage forms are prepared that include an effective amount of melt-extruded multiparticulates within a capsule. For example, a plurality of the melt-extruded multiparticulates may be placed in a gelatin capsule in an amount sufficient to provide an effective controlled release dose when ingested and contacted by gastric fluid.


In another preferred embodiment, a suitable amount of the multiparticulate extrudate is compressed into an oral tablet using conventional tableting equipment using standard techniques. Techniques and compositions for making tablets (compressed and molded), capsules (hard and soft gelatin) and pills are also described in Remington's Pharmaceutical Sciences, (Arthur Osol, editor), 1553-1593 (1980), incorporated by reference herein.


In yet another preferred embodiment, the extrudate can be shaped into tablets as set forth in U.S. Pat. No. 4,957,681 (Klimesch, et. al.), described in additional detail above and hereby incorporated by reference.


Optionally, the controlled-release matrix multiparticulate systems or tablets can be coated, or the gelatin capsule can be further coated, with a controlled release coating such as the controlled release coatings described above. Such coatings preferably include a sufficient amount of hydrophobic and/or hydrophilic controlled-release material to obtain a weight gain level from about 2 to about 25 percent, although the overcoat may be greater depending upon, e.g., the physical properties of the particular opioid analgesic used and the desired release rate, among other things.


The dosage forms of the present invention may further include combinations of melt-extruded multiparticulates containing one or more opioid analgesics. Furthermore, the dosage forms can also include an amount of an immediate release therapeutically active agent for prompt therapeutic effect. The immediate release therapeutically active agent may be incorporated, e.g., as separate pellets within a gelatin capsule, or may be coated on the surface of, e.g., beads or melt extruded multiparticulates. The unit dosage forms of the present invention may also contain a combination of, e.g., controlled release beads and matrix multiparticulates to achieve a desired effect.


The controlled-release formulations of the present invention preferably slowly release the therapeutically active agent, e.g., when ingested and exposed to gastric fluids, and then to intestinal fluids. The controlled-release profile of the melt-extruded formulations of the invention can be altered, for example, by varying the amount of controlled-release material, by varying the amount of plasticizer relative to other matrix constituents, hydrophobic material, by the inclusion of additional ingredients or excipients, by altering the method of manufacture, etc.


In other embodiments of the invention, melt-extruded formulations are prepared without the inclusion of the therapeutically active agent, which is added thereafter to the extrudate. Such formulations typically will have the therapeutically active agent blended together with the extruded matrix material, and then the mixture would be tableted in order to provide a slow release formulation. Such formulations may be advantageous, for example, when the therapeutically active agent included in the formulation is sensitive to temperatures needed for softening the hydrophobic material and/or the retardant material.


Typical melt-extrusion production systems suitable for use in accordance with the present invention include a suitable extruder drive motor having variable speed and constant torque control, start-stop controls, and ammeter. In addition, the production system will include a temperature control console which includes temperature sensors, cooling means and temperature indicators throughout the length of the extruder. In addition, the production system will include an extruder such as twin-screw extruder which consists of two counter-rotating intermeshing screws enclosed within a cylinder or barrel having an aperture or die at the exit thereof. The feed materials enter through a feed hopper and are moved through the barrel by the screws and are forced through the die into strands which are thereafter conveyed such as by a continuous movable belt to allow for cooling and being directed to a pelletizer or other suitable device to render the extruded ropes into the multiparticulate system. The pelletizer can consist of rollers, fixed knife, rotating cutter and the like. Suitable instruments and systems are available from distributors such as C.W. Brabender Instruments, Inc. of South Hackensack, N.J. Other suitable apparatus will be apparent to those of ordinary skill in the art.


A further aspect of the invention is related to the preparation of melt-extruded multiparticulates as set forth above in a manner which controls the amount of air included in the extruded product. By controlling the amount of air included in the extrudate, it has been surprisingly found that the release rate of the therapeutically active agent from the, e.g., multiparticulate extrudate, can be altered significantly. In certain embodiments, it has been surprisingly found that the pH dependency of the extruded product can be altered as well.


Thus, in a further aspect of the invention, the melt-extruded product is prepared in a manner which substantially excludes air during the extrusion phase of the process. This may be accomplished, for example, by using a Leistritz extruder having a vacuum attachment. It has been surprisingly found that extruded multiparticulates prepared according to the invention using the Leistritz extruder under vacuum provides a melt-extruded product having different physical characteristics. In particular, the extrudate is substantially non-porous when magnified, e.g., using a scanning electron microscope which provides an SEM (scanning electron micrograph). Contrary to conventional thought, it has been found that such substantially non-porous formulations provide a faster release of the therapeutically active agent, relative to the same formulation prepared without vacuum. SEMs of the multiparticulates prepared using an extruder under vacuum appear very smooth, and the multiparticulates tend to be more robust than those multiparticulates prepared without vacuum. It has been observed that in at least certain formulations, the use of extrusion under vacuum provides an extruded multiparticulate product which is more pH-dependent than its counterpart formulation prepared without vacuum.


Processes for Preparing Matrix Beads

Controlled-release dosage forms according to the present invention may also be prepared as matrix beads formulations. The matrix beads include a spheronising agent and the hydrocodone.


The hydrocodone preferably comprises from about 0.01 to about 99% by weight of the matrix bead by weight. It is preferable that the hydrocodone is included as about 0.1 to about 50% by weight of the matrix bead.


Spheronising agents which may be used to prepare the matrix bead formulations of the present invention include any art-known spheronising agent. Cellulose derivatives are preferred, and microcrystalline cellulose is especially preferred. A suitable microcrystalline cellulose is, for example, the material sold as Avicel PH 101 (Trade Mark, FMC Corporation). The spheronising agent is preferably included as about 1 to about 99% of the matrix bead by weight.


In addition to the active ingredient and spheronizing agent, the spheroids may also contain a binder. Suitable binders, such as low viscosity, water soluble polymers, will be well known to those skilled in the pharmaceutical art. However, water soluble hydroxy lower alkylcellulose, such as hydroxypropylcellulose, are preferred.


In addition to the opioid analgesic and spheronising agent, the matrix bead formulations of the present invention may include a controlled release material such as those described hereinabove. Preferred controlled-release materials for inclusion in the matrix bead formulations include acrylic and methacrylic acid polymers or copolymers, and ethylcellulose. When present in the formulation, the controlled-release material will be included in amounts of from about 1 to about 80% of the matrix bead, by weight. The controlled-release material is preferably included in the matrix bead formulation in an amount effective to provide controlled release of the opioid analgesic from the bead.


Pharmaceutical processing aids such as binders, diluents, and the like may be included in the matrix bead formulations. Amounts of these agents included in the formulations will vary with the desired effect to be exhibited by the formulation.


The matrix beads may be overcoated with a controlled-release coating including a controlled-release material such as those described hereinabove. The controlled-release coating is applied to a weight gain of from about 5 to about 30%. The amount of the controlled-release coating to be applied will vary according to a variety of factors, e.g., the composition of the matrix bead and the chemical and/or physical properties of the opioid analgesic (i.e., hydrocodone).


Matrix beads are generally prepared by granulating the spheronising agent together with the opioid analgesic, e.g. by wet granulation. The granulate is then spheronized to produce the matrix beads. The matrix beads are then optionally overcoated with the controlled release coating by methods such as those described hereinabove.


Another method for preparing matrix beads, for example, by (a) forming granules comprising at least one water soluble hydroxyalkyl cellulose and opioid or an opioid salt; (b) mixing the hydroxyalkyl cellulose containing granules with at least one C12-C36 aliphatic alcohol; and (c) optionally, compressing and shaping the granules. Preferably, the granules are formed by wet granulating the hydroxyalkyl cellulose/opioid with water. In a particularly preferred embodiment of this process, the amount of water added during the wet granulation step is preferably between 1.5 and 5 times, especially between 1.75 and 3.5 times, the dry weight of the opioid.


In yet other alternative embodiments, a spheronizing agent, together with the active ingredient can be spheronized to form spheroids. Microcrystalline cellulose is preferred. A suitable microcrystalline cellulose is, for example, the material sold as Avicel PH 101 (Trade Mark, FMC Corporation). In such embodiments, in addition to the active ingredient and spheronizing agent, the spheroids may also contain a binder. Suitable binders, such as low viscosity, water soluble polymers, will be well known to those skilled in the pharmaceutical art. However, water soluble hydroxy lower alkyl cellulose, such as hydroxy propyl cellulose, are preferred. Additionally (or alternatively) the spheroids may contain a water insoluble polymer, especially an acrylic polymer, an acrylic copolymer, such as a methacrylic acid-ethyl acrylate copolymer, or ethyl cellulose. In such embodiments, the sustained-release coating will generally include a water insoluble material such as (a) a wax, either alone or in admixture with a fatty alcohol; or (b) shellac or zein.


Controlled Release Bead Formulations

In one especially preferred embodiment, the oral dosage form comprises an effective number of controlled release spheroids contained within a gelatin capsule.


In another preferred embodiment of the present invention, the controlled-release dosage form comprises spheroids containing the active ingredient coated with a controlled-release coating including a controlled release material. The term spheroid is known in the pharmaceutical art and means, e.g., a spherical granule having a diameter of between 0.1 mm and 2.5 mm, especially between 0.5 mm and 2 mm.


The spheroids are preferably film coated with a controlled release material that permits release of the opioid (or salt) at a controlled rate in an aqueous medium. The film coat is chosen so as to achieve, in combination with the other stated properties, the in-vitro release rate outlined above (e.g., at least about 12.5% released after 1 hour). The controlled-release coating formulations of the present invention preferably produce a strong, continuous film that is smooth and elegant, capable of supporting pigments and other coating additives, non-toxic, inert, and tack-free.


Coatings

The dosage forms of the present invention may optionally be coated with one or more coatings suitable for the regulation of release or for the protection of the formulation. In one embodiment, coatings are provided to permit either pH-dependent or pH-independent release, e.g., when exposed to gastrointestinal fluid. When a pH-independent coating is desired, the coating is designed to achieve optimal release regardless of pH-changes in the environmental fluid, e.g., the GI tract. Other preferred embodiments include a pH-dependent coating that releases the opioid in desired areas of the gastro-intestinal (GI) tract, e.g., the stomach or small intestine, such that an absorption profile is provided which is capable of providing at least about twelve hour and preferably up to twenty-four hour analgesia to a patient. It is also possible to formulate compositions which release a portion of the dose in one desired area of the GI tract, e.g., the stomach, and release the remainder of the dose in another area of the GI tract, e.g., the small intestine.


Formulations according to the invention that utilize pH-dependent coatings may also impart a repeat-action effect whereby unprotected drug is coated over an enteric coat and is released in the stomach, while the remainder, being protected by the enteric coating, is released further down the gastrointestinal tract. Coatings which are pH-dependent may be used in accordance with the present invention include a controlled release material such as, e.g., shellac, cellulose acetate phthalate (CAP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose phthalate, and methacrylic acid ester copolymers, zein, and the like.


In another preferred embodiment, the present invention is related to a stabilized solid controlled dosage form comprising an opioid coated with a hydrophobic controlled release material selected from (i) an alkylcellulose; (ii) an acrylic polymer, or (iii) mixtures thereof. The coating may be applied in the form of an organic or aqueous solution or dispersion.


In certain preferred embodiments, the controlled release coating is derived from an aqueous dispersion of the hydrophobic controlled release material. The coated substrate containing the opioid(s) (e.g., a tablet core or inert pharmaceutical beads or spheroids) is then cured until an endpoint is reached at which the substrate provides a stable dissolution. The curing endpoint may be determined by comparing the dissolution profile (curve) of the dosage form immediately after curing to the dissolution profile (curve) of the dosage form after exposure to accelerated storage conditions of, e.g., at least one month at a temperature of 40° C. and a relative humidity of 75%. These formulations are described in detail in U.S. Pat. Nos. 5,273,760 and 5,286,493, assigned to the Assignee of the present invention and hereby incorporated by reference. Other examples of controlled-release formulations and coatings which may be used in accordance with the present invention include Assignee's U.S. Pat. Nos. 5,324,351; 5,356,467, and 5,472,712, hereby incorporated by reference in their entirety.


In preferred embodiments, the controlled release coatings include a plasticizer such as those described herein below.


In certain embodiments, it is necessary to overcoat the substrate comprising the opioid analgesic with a sufficient amount of the aqueous dispersion of e.g., alkylcellulose or acrylic polymer, to obtain a weight gain level from about 2 to about 50%, e.g., about 2 to about 25% in order to obtain a controlled-release formulation. The overcoat may be lesser or greater depending upon the physical properties of the therapeutically active agent and the desired release rate, the inclusion of plasticizer in the aqueous dispersion and the manner of incorporation of the same, for example.


Alkylcellulose Polymers

Cellulosic materials and polymers, including alkylcelluloses are controlled release materials well suited for coating the substrates, e.g., beads, tablets, etc. according to the invention. Simply by way of example, one preferred alkylcellulosic polymer is ethylcellulose, although the artisan will appreciate that other cellulose and/or alkylcellulose polymers may be readily employed, singly or on any combination, as all or part of a hydrophobic coatings according to the invention.


One commercially-available aqueous dispersion of ethylcellulose is Aquacoat® (FMC Corp., Philadelphia, Pa., U.S.A.). Aquacoat® is prepared by dissolving the ethylcellulose in a water-immiscible organic solvent and then emulsifying the same in water in the presence of a surfactant and a stabilizer. After homogenization to generate submicron droplets, the organic solvent is evaporated under vacuum to form a pseudolatex. The plasticizer is not incorporated in the pseudolatex during the manufacturing phase. Thus, prior to using the same as a coating, it is necessary to intimately mix the Aquacoat® with a suitable plasticizer prior to use.


Another aqueous dispersion of ethylcellulose is commercially available as Surelease® (Colorcon, Inc., West Point, Pa., U.S.A.). This product is prepared by incorporating plasticizer into the dispersion during the manufacturing process. A hot melt of a polymer, plasticizer (dibutyl sebacate), and stabilizer (oleic acid) is prepared as a homogeneous mixture, which is then diluted with an alkaline solution to obtain an aqueous dispersion which can be applied directly onto substrates.


Acrylic Polymers

In other preferred embodiments of the present invention, the controlled release material comprising the controlled-release coating is a pharmaceutically acceptable acrylic polymer, including but not limited to acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), polymethacrylate, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, poly(methacrylic acid anhydride), and glycidyl methacrylate copolymers.


In certain preferred embodiments, the acrylic polymer is comprised of one or more ammonio methacrylate copolymers. Ammonio methacrylate copolymers are well known in the art, and are described in NF XVII as fully polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammonium groups.


In order to obtain a desirable dissolution profile, it may be necessary to incorporate two or more ammonio methacrylate copolymers having differing physical properties, such as different molar ratios of the quaternary ammonium groups to the neutral (meth)acrylic esters.


Certain methacrylic acid ester-type polymers are useful for preparing pH-dependent coatings which may be used in accordance with the present invention. For example, there are a family of copolymers synthesized from diethylaminoethyl methacrylate and other neutral methacrylic esters, also known as methacrylic acid copolymer or polymeric methacrylates, commercially available as Eudragit® from Röhm Tech, Inc. There are several different types of Eudragit®. For example, Eudragit E is an example of a methacrylic acid copolymer which swells and dissolves in acidic media. Eudragit L is a methacrylic acid copolymer which does not swell at about pH<5.7 and is soluble at about pH>6. Eudragit S does not swell at about pH<6.5 and is soluble at about pH>7. Eudragit RL and Eudragit RS are water swellable, and the amount of water absorbed by these polymers is pH-dependent, however, dosage forms coated with Eudragit RL and RS are pH-independent.


In certain preferred embodiments, the acrylic coating comprises a mixture of two acrylic resin lacquers commercially available from Rohm Pharma under the Tradenames Eudragit® RL30D and Eudragit® RS30D, respectively. Eudragit® RL30D and Eudragit® RS30D 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® RL30D and 1:40 in Eudragit® RS30D. The mean molecular weight is about 150,000. The code designations RL (high permeability) and RS (low permeability) refer to the permeability properties of these agents. Eudragit® RL/RS mixtures are insoluble in water and in digestive fluids. However, coatings formed from the same are swellable and permeable in aqueous solutions and digestive fluids.


The Eudragit® RL/RS dispersions of the present invention may be mixed together in any desired ratio in order to ultimately obtain a controlled-release formulation having a desirable dissolution profile. Desirable controlled-release formulations may be obtained, for instance, from a retardant coating derived from 100% Eudragit® RL, 50% Eudragit® RL and 50% Eudragit® RS, and 10% Eudragit® RL:Eudragit® 90% RS. Of course, one skilled in the art will recognize that other acrylic polymers may also be used, such as, for example, Eudragit® L.


Plasticizers

In embodiments of the present invention where the coating comprises an aqueous dispersion of a hydrophobic controlled release material, the inclusion of an effective amount of a plasticizer in the aqueous dispersion of hydrophobic material will further improve the physical properties of the controlled-release coating. For example, because ethylcellulose has a relatively high glass transition temperature and does not form flexible films under normal coating conditions, it is preferable to incorporate a plasticizer into an ethylcellulose coating containing controlled-release coating before using the same as a coating material. Generally, the amount of plasticizer included in a coating solution is based on the concentration of the film-former, e.g., most often from about 1 to about 50 percent by weight of the film-former. Concentration of the plasticizer, however, can only be properly determined after careful experimentation with the particular coating solution and method of application.


Examples of suitable plasticizers for ethylcellulose include water insoluble plasticizers such as dibutyl sebacate, diethyl phthalate, triethyl citrate, tibutyl citrate, and triacetin, although it is possible that other water-insoluble plasticizers (such as acetylated monoglycerides, phthalate esters, castor oil, etc.) may be used. Triethyl citrate is an especially preferred plasticizer for the aqueous dispersions of ethyl cellulose of the present invention.


Examples of suitable plasticizers for the acrylic polymers of the present invention include, but are not limited to citric acid esters such as triethyl citrate NF XVI, tributyl citrate, dibutyl phthalate, and possibly 1,2-propylene glycol. Other plasticizers which have proved to be suitable for enhancing the elasticity of the films formed from acrylic films such as Eudragit® RL/RS lacquer solutions include polyethylene glycols, propylene glycol, diethyl phthalate, castor oil, and triacetin. Triethyl citrate is an especially preferred plasticizer for the aqueous dispersions of ethyl cellulose of the present invention.


It has further been found that the addition of a small amount of talc to the controlled release coating reduces the tendency of the aqueous dispersion to stick during processing, and acts as a polishing agent.


Preparation of Coated Bead Formulations

When an aqueous dispersion of hydrophobic material is used to coat substrates, e.g., inert pharmaceutical beads such as nu pariel 18/20 beads, a plurality of the resultant stabilized solid controlled-release beads may thereafter be placed in a gelatin capsule in an amount sufficient to provide an effective controlled-release dose when ingested and contacted by an environmental fluid, e.g., gastric fluid or dissolution media.


The stabilized controlled-release bead formulations of the present invention slowly release the opioid analgesic, e.g., when ingested and exposed to gastric fluids, and then to intestinal fluids. The controlled-release profile of the formulations of the invention can be altered, for example, by varying the amount of overcoating with the aqueous dispersion of hydrophobic controlled release material, altering the manner in which the plasticizer is added to the aqueous dispersion of hydrophobic controlled release material, by varying the amount of plasticizer relative to hydrophobic controlled release material, by the inclusion of additional ingredients or excipients, by altering the method of manufacture, etc. The dissolution profile of the ultimate product may also be modified, for example, by increasing or decreasing the thickness of the controlled release coating.


Substrates coated with a therapeutically active agent are prepared, e.g. by dissolving the therapeutically active agent in water and then spraying the solution onto a substrate, for example, nu pariel 18/20 beads, using a Wuster insert. Optionally, additional ingredients are also added prior to coating the beads in order to assist the binding of the opioid to the beads, and/or to color the solution, etc. For example, a product which includes hydroxypropyl methylcellulose, etc. with or without colorant (e.g., Opadry®, commercially available from Colorcon, Inc.) may be added to the solution and the solution mixed (e.g., for about 1 hour) prior to application of the same onto the substrate. The resultant coated substrate may then be optionally overcoated with a barrier agent, to separate the therapeutically active agent from the hydrophobic controlled-release coating.


An example of a suitable barrier agent is one which comprises hydroxypropyl methylcellulose. However, any film-former known in the art may be used. It is preferred that the barrier agent does not affect the dissolution rate of the final product.


The substrates may then be overcoated with an aqueous dispersion of the hydrophobic controlled release material. The aqueous dispersion of hydrophobic controlled release material preferably further includes an effective amount of plasticizer, e.g. triethyl citrate. Pre-formulated aqueous dispersions of ethylcellulose, such as Aquacoat® or Surelease®, may be used. If Surelease® is used, it is not necessary to separately add a plasticizer. Alternatively, pre-formulated aqueous dispersions of acrylic polymers such as Eudragit® can be used.


The coating solutions of the present invention preferably contain, in addition to the film-former, plasticizer, and solvent system (i.e., water), a colorant to provide elegance and product distinction. Color may be added to the solution of the therapeutically active agent instead, or in addition to the aqueous dispersion of hydrophobic material. For example, color can be added to Aquacoat® via the use of alcohol or propylene glycol based color dispersions, milled aluminum lakes and opacifiers such as titanium dioxide by adding color with shear to water soluble polymer solution and then using low shear to the plasticized Aquacoat®. Alternatively, any suitable method of providing color to the formulations of the present invention may be used. Suitable ingredients for providing color to the formulation when an aqueous dispersion of an acrylic polymer is used include titanium dioxide and color pigments, such as iron oxide pigments. The incorporation of pigments, may, however, increase the retard effect of the coating.


The plasticized aqueous dispersion of hydrophobic controlled release material may be applied onto the substrate comprising the therapeutically active agent by spraying using any suitable spray equipment known in the art. In a preferred method, a Wurster fluidized-bed system is used in which an air jet, injected from underneath, fluidizes the core material and effects drying while the acrylic polymer coating is sprayed on. A sufficient amount of the aqueous dispersion of hydrophobic material to obtain a predetermined controlled-release of said therapeutically active agent when said coated substrate is exposed to aqueous solutions, e.g. gastric fluid, is preferably applied, taking into account the physical characteristics of the therapeutically active agent, the manner of incorporation of the plasticizer, etc. After coating with the hydrophobic controlled release material, a further overcoat of a film-former, such as Opadry®, is optionally applied to the beads. This overcoat is provided, if at all, in order to substantially reduce agglomeration of the beads.


The release of the therapeutically active agent from the controlled-release formulation of the present invention can be further influenced, i.e., adjusted to a desired rate, by the addition of one or more release-modifying agents, or by providing one or more passageways through the coating. The ratio of hydrophobic controlled release material to water soluble material is determined by, among other factors, the release rate required and the solubility characteristics of the materials selected.


The release-modifying agents which function as pore-formers may be organic or inorganic, and include materials that can be dissolved, extracted or leached from the coating in the environment of use. The pore-formers may comprise one or more hydrophilic materials such as hydroxypropylmethylcellulose.


The controlled-release coatings of the present invention can also include erosion-promoting agents such as starch and gums.


The controlled-release coatings of the present invention can also include materials useful for making microporous lamina in the environment of use, such as polycarbonates comprised of linear polyesters of carbonic acid in which carbonate groups reoccur in the polymer chain.


The release-modifying agent may also comprise a semi-permeable polymer. In certain preferred embodiments, the release-modifying agent is selected from hydroxypropylmethylcellulose, lactose, metal stearates, and mixtures of any of the foregoing.


The controlled-release coatings of the present invention may also include an exit means comprising at least one passageway, orifice, or the like. The passageway may be formed by such methods as those disclosed in U.S. Pat. Nos. 3,845,770; 3,916,889; 4,063,064; and 4,088,864, all of which are hereby incorporated by reference. The passageway can have any shape such as round, triangular, square, elliptical, irregular, etc.


Another method of producing controlled release bead formulations suitable for about 24-hour administration is via powder layering. U.S. Pat. No. 5,411,745, assigned to the Assignee of the present invention and hereby incorporated by reference in its entirety, teaches preparation of 24-hour morphine formulations prepared via powder layering techniques utilizing a processing aid consisting essentially of hydrous lactose impalpable. The powder-layered beads are prepared by spraying an aqueous binder solution onto inert beads to provide a tacky surface, and subsequently spraying a powder that is a homogenous mixture of morphine sulfate and hydrous lactose impalpable onto the tacky beads. The beads are then dried and coated with a hydrophobic material such as those described hereinabove to obtain the desired release of drug when the final formulation is exposed to environmental fluids. An appropriate amount of the controlled release beads are then, e.g. encapsulated to provide a final dosage form which provides effective plasma concentrations of morphine for about 12 hours.


DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS

The following examples illustrate various aspects of the present invention. They are not meant to be construed to limit the claims in any manner whatsoever.


Example 1

Hydrocodone sustained release tablets were produced with the formula set forth in Table 1 below:












TABLE 1






Ingredients
Amt/Unit (mg)
Amount/Batch (gm)



















Hydrocodone Bitartrate
15.0
150.0



Spray Dried Lactose
56.0
560.0



Povidone
4.0
40.0



Eudragit RS30D (solids)
10.0
100.0



Triacetin
2.0
20.0



Stearyl Alcohol
20.0
200.0



Talc
2.0
20.0



Magnesium Stearate
1.0
10.0



Total
110.0
1100.0









According to the following procedure:

  • 1. Retardant dispersion: Blend Eudragit RS30D and Triacetin using a lightning mixer.
  • 2. Melt Stearyl Alcohol.
  • 3. Spray retardant dispersion onto Hydrocodone Bitartrate, Spray Dried Lactose, and Povidone using a fluid bed granulator.
  • 4. Dry batch on a stainless steel tray for 15 minutes, or till constant weight.
  • 5. Incorporate the melted Stearyl Alcohol into the batch using a Hobart mixer.
  • 6. Dry waxed granulation on a stainless steel tray for 30 minutes, or temperature of granulation reaches 35° C. or less.
  • 7. Mill the cooled granulation through a CoMil.
  • 8. Lubricate the granulation with talc and magnesium stearate using a Hobart Mixer.
  • 9. Compress the granulation into tablets using a tablet press.


The tablets were then tested for dissolution using the following procedure:

  • 1. Apparatus: USP Method I (basket), 100 rpm.
  • 2. Medium: 700 ml SGF for 55 min, thereafter 900 ml of SIF without enzyme
  • 3. Sampling time: 1, 2, 4, 8 and 12 hours.
  • 4. Analytical: High Performance Liquid Chromatography.


The dissolution parameters are set forth in Table II below:











TABLE II






Time (Hours)
% Dissolved


















1
39.7



2
51.5



4
67.4



8
86.4



12
96.1









The Cmax and Tmax were then obtained for Example 1 and an immediate release reference standard in a bioavailability study comparing hydrocodone 15 mg administered as an immediate release formulation (Lortab 7.5 mg×2) to the above CR formulation in healthy human subjects, as set forth in Table I below:











TABLE III






Pharmacokinetic data
Hydrocodone Bitartrate


















Cmax (ng/ml)
35.4



IR reference product




Cmax (ng/ml)
13.4



CR product




Cmax (CR)/Cmax (IR)
38%



Tmax (hr)
1.32



IR reference product




Tmax (hr)
4.07



CR product









Example 2

Hydrocodone sustained release tablets were produced with the formula set forth in Table IV below:












TABLE IV






Ingredients
Amt/Unit (mg)
Amt/Batch (g)



















Hydrocodone Bitartrate
15.0
150.0



Spray Dried Lactose
51.0
510.0



Povidone
4.0
40.0



Eudragit RS30D
10.0
100.0



(solids)





Triacetin
2.0
20.0



Stearyl Alcohol
25.0
250.0



Talc
2.0
20.0



Magnesium Stearate
1.0
10.0



Total
110.0
1100.0









according to the procedure of Example 1.


The dissolution parameters were then obtained using the procedure of Example 1. The results are set forth in table V below:











TABLE V






Time (Hours)
% Dissolved


















1
36



2
45.8



4
60.5



8
78.9



12
90.4









Example 3

Hydrocodone sustained release capsules were produced with the formula set forth in Table VI below:












TABLE VI






Ingredients
Amt/Unit (mg)
Amt/Batch (g)



















Hydrocodone
15.0
320.0



Bitartrate





Eudragit RSPO
76.0
1520.0



Eudragit RLPO
4.0
80.0



Stearyl Alcohol
25.0
500.0



Total
120.0
2400.0









According to the following procedure:

  • 1. Blend milled Stearyl Alcohol, Eudragit RLPO, Hydrocodone Bitartrate, and Eudragit RSPO using a Hobart Mixer.
  • 2. Extrude the granulation using a Powder Feeder, Melt Extruder (equipped with the 6×1 mm die head), Conveyor, Lasermike, and Pelletizer under the following conditions:

















Zone 1
 10° C.



Zone 2
 20° C.



Zone 3
120° C.



Zone 4
120° C.



Zone 5
120° C.



Zone 6
120° C.



Zone 7
 95° C.



Zone 8
 95° C.



MGA
120° C.



Die
117° C.










Powder feed rate—40 g/min; screw speed—185 rpm; vacuum—˜980 mBar


Conveyor—such that diameter of extrudate is 1 mm


Pelletizer—such that pellets are cut to 1 mm in length
  • 3. Screen pellets using #16 mesh and #20 mesh screens. Collect material that passes through the #16 mesh screen and is retained on the #20 mesh screen.
  • 4. Fill size #2 clear gelatin capsules with the pellets. Range: NLT 114 mg and NMT 126 mg.


The dissolution parameters were then obtained using the procedure of Example 1. The results are set forth in table VII below:











TABLE VII






Time (Hours)
% Dissolved


















1
23.9



2
34.7



4
51.7



8
74.6



12
85.2









Example 4

Oxycodone sustained release tablets were produced with the formula set forth in Table VIII below:











TABLE VIII





Ingredients
Amt/Unit (mg)
Amount/Batch (gm)

















Oxycodone HCl
20.0
22.0


Spray Dried Lactose
59.25
65.175


Povidone
5.0
5.5


Eudragit RS30D (solids)
10.0
11.0


Triacetin
2.0
2.2


Stearyl Alcohol
25.0
27.5


Talc
2.5
2.75


Magnesium Stearate
1.25
1.375


Opadry Pink Y-S-14518A
4.0
4.26


Total
129.0
141.76









According to the following procedure:

  • 1. Granulation: Spray the Eudragit/Triacetin dispersion onto the Oxycodone HCl, Spray Dried Lactose and Povidone using a fluid bed granulator.
  • 2. Milling: Discharge the granulation and pass through a mill.
  • 3. Waxing: Melt the stearyl alcohol and add to the milled granulation using a mixer. Allow to cool.
  • 4. Milling: Pass the cooled granulation through a mill.
  • 5. Lubrication: Lubricate the granulation with talc and magnesium stearate using a mixer.
  • 6. Compression: Compress the granulation into tablets using a tablet press.
  • 7. Film coating: Apply an aqueous film coat to the tablets.


The tablets were then tested for dissolution using the following procedure:

  • 1. Apparatus: USP Type II (paddle), 150 rpm.
  • 2. Medium: 700 ml SGF for first hour, thereafter made 900 ml with phosphate buffer to pH 7.5.
  • 3. Sampling time: 1, 2, 4, 8, 12, 18 and 24 hours.
  • 4. Analytical: High Performance Liquid Chromatography.


The dissolution parameters are set forth in Table IX below:











TABLE IX






Time (hrs)
% Dissolved


















1
45



2
55



4
70



8
87



12
96



18
101



24
102









The Cmax and Tmax were then obtained for Example 4 and an immediate release reference standard in a bioavailability study, as set forth in Table X below:











TABLE X






Pharmacokinetic data
Oxycodone HCl


















Cmax (ng/ml)
38.2



IR reference product




Cmax (ng/ml)
21.7



CR product




Cmax (CR)/Cmax (IR)
57%



Tmax (hr)
1.10



IR reference product




Tmax (hr)
2.62



CR product









Example 5

Morphine sustained release tablets were produced with the formula set forth in Table XI below:












TABLE XI







Amount/unit
Amount/batch



Ingredients
(mg)
(kg)



















Morphine sulfate
30.0
138.0



Spray Dried Lactose
70.0
322.0



Hydroxyethyl cellulose
10.0
46.0



Cetostearyl alcohol
35.0
161.0



Talc
3.0
13.8



Magnesium stearate
2.0
9.2



Opadry YS-1-4729
5.0
23.0



Total
155.0
713.0









According to the following procedure:

  • 1. Granulation: Add water to the Morphine sulfate, Spray Dried Lactose and Hydroxyethyl cellulose in a mixer and dry using a fluid bed granulator.
  • 2. Screening: Discharge the granulation and pass through a sieve.
  • 3. Waxing: Melt the cetostearyl alcohol and add to the milled granulation using a mixer. Allow to cool.
  • 4. Screening: Pass the cooled granulation through a sieve.
  • 5. Lubrication: Lubricate the granulation with talc and magnesium stearate using a mixer.
  • 6. Compression: Compress the granulation into tablets using a tablet press.
  • 7. Film coating: Apply an aqueous film coat to the tablets.


The tablets were then tested for dissolution using the following procedure:

  • 1. Apparatus: USP Method I (Basket), 50 rpm
  • 2. Medium: 900 ml of Purified Water, 37° C.
  • 3. Sampling time: 1, 2, 3, 4, and 6 hours.
  • 4. Analytical: UV detection, 285 nm and 305 nm, 2-point method using 5-cm cell.


The dissolution parameters are set forth in Table XII below:











TABLE XII






Time (Hours)
% Dissolved








1
34.2



2
49.9



3
64.2



4
75.5



6
90.3









The Cmax and Tmax were then obtained for Example 5 and an immediate release reference standard in a bioavailability study, as set forth in Table XIII below:











TABLE XIII






Pharmacokinetic data
Morphine Sulphate


















Cmax (ng/ml)
22.1



IR reference product




Cmax (ng/ml)
12



CR product




Cmax (CR)/Cmax (IR)
54%



Tmax (hr)
0.98



IR reference product




Tmax (hr)
2.09



CR product









Example 6

The pharmacokinetic parameters of Example 1, Example 4 and Example 5 were compared to each other. It was surprisingly found that even though the dissolution of the hydrocodone HCl controlled release tablets of example 1 were very similar to the dissolution of the controlled release oxycodone tablets of example 4 and the morphine sulfate controlled release tablets of example 5, the Cmax ratio of CR to IR for the hydrocodone formulation is 38%, whereas the oxycodone tablets and morphine tablets are over 50%. The comparative results are set forth in Table XIV below:












TABLE XIV






Hydrocodone
Oxycodone
Morphine


Pharmacokinetic data
Bitartrate
HCl
Sulphate


















Cmax (ng/ml)
35.4
38.2
22.1


IR reference product





Cmax (ng/ml)
13.4
21.7
12


CR product





Cmax (CR)/Cmax (IR)
38%
57%
54%


Tmax (hr)
1.32
1.10
0.98


IR reference product





Tmax (hr)
4.07
2.62
2.09


CR product









Example 7

A single dose, four treatment, open label, pharmacokinetic comparison of controlled release hydrocodone formulations of Example 1, Example 2, Example 3 and two immediate release hydrocodone bitartrate 7.5 mg/Acetaminophen 500 mg tablets (IR Example) in fasted normal volunteers was conducted. The plasma concentrations for these formulations are set forth in tables 15-18 below:









TABLE 15







Hydrocodone Plasma Concentration (ng/mL) after administration of one (1)


Controlled-Release Hydrocodone Bitartrate 15 mg tablet-Formulation A









Time (hours)





















Subject
−0.08
0.5
0.75
1
2
3
4
6
9
12
18
24
30
36
























1
0.00
4.55
11.1
9.11
15.8
15.5
17.4
15.4
14.5
12.1
6.33
3.58
2.25
1.29


2
0.00
7.81
8.76
9.20
11.3
14.8
15.5
14.5
10.5
9.30
5.40
3.39
2.10
0.921


3
0.00
4.63
7.66
8.95
15.9
15.6
16.9
16.3
12.3
9.41
6.55
4.10
2.38
0.986


4
0.00
3.48
9.48
9.11
10.7
11.9
13.0
12.4
10.7
8.96
5.22
3.08
1.56
0.558


5
0.00
1.43
4.25
7.20
12.8
13.5
13.0
12.5
9.62
7.01
4.38
3.26
1.93
1.01


6
0.00
4.69
7.60
10.5
12.8
13.9
13.3
15.1
12.3
8.59
4.52
3.11
1.59
0.702


7
0.00
0.56
1.86
3.85
7.54
8.26
8.18
8.90
6.23
4.56
2.99
1.61
0.752
0.00


8
0.00
3.68
7.61
11.5
12.4
13.2
12.7
12.5
9.10
7.09
4.33
2.93
1.24
0.509


9
0.00
8.06
9.79
9.98
11.4
10.7
11.4
11.9
7.66
5.98
3.85
2.10
1.12
0.573


10
0.00
3.83
5.71
7.84
8.49
10.8
11.6
11.5
8.02
6.70
3.34
2.33
1.31
0.00


11
0.00
3.64
5.20
8.00
10.3
11.8
12.5
10.8
7.44
7.84
4.75
2.21
1.11
0.00


12
0.00
3.07
6.14
8.51
14.3
15.0
14.9
14.7
12.1
7.75
4.34
2.52
1.69
0.859


13
0.00
1.95
3.82
4.47
9.55
9.15
8.31
8.05
5.85
3.93
2.45
7.68
1.35
1.07


14
0.00
2.21
4.56
7.33
11.2
12.9
13.3
13.2
10.6
8.41
4.68
3.11
2.35
0.978


MEAN
0.00
3.83
6.68
8.25
11.7
12.6
13.0
12.7
9.78
7.69
4.51
3.22
1.62
0.675


SD
0.00
2.13
2.62
2.10
2.48
2.31
2.70
2.41
2.54
2.09
1.15
1.44
0.513
0.425


% CV
0.00
21.7
39.2
25.5
21.2
18.3
20.8
19.0
26.0
27.2
25.5
44.7
31.7
63.0
















TABLE 16







Hydrocodone Plasma Concentration (ng/mL) after administration of one (1)


Controlled-Release Hydrocodone Bitartrate 15 mg tablet-Formulation B









Time (hours)





















Subject
−0.08
0.5
0.75
1
2
3
4
6
9
12
18
24
30
36
























1
0.00
3.18
5.64
11.8
11.4
12.4
13.5
14.3
11.4
9.28
5.69
3.23
2.23
1.10


2
0.00
2.61
7.04
8.53
10.7
12.4
11.5
13.6
11.4
9.25
6.43
4.13
2.59
1.35


3
0.00
5.49
7.57
9.67
13.5
15.6
15.7
14.4
12.6
9.41
7.83
5.19
3.45
1.77


4
0.00
2.71
5.67
6.35
8.88
11.3
13.7
12.0
8.72
8.18
5.58
4.33
2.63
1.26


5
0.00
3.98
6.59
7.38
10.6
11.8
11.6
9.42
6.75
4.81
5.28
3.67
2.43
1.25


6
0.00
0.711
2.85
7.98
12.9
13.6
13
13.8
10.1
8.04
5.17
3.71
2.33
0.940


7
0.00
1.82
3.03
3.97
7.22
8.04
8.05
7.87
5.97
3.77
2.53
2.12
1.94
1.19


8
0.00
2.47
3.99
6.03
10.9
13.2
13.8
12.6
9.49
7.60
6.11
4.74
2.38
0.856


9
0.00
5.02
10.4
8.48
9.06
9.90
9.88
7.96
4.78
3.99
3.77
3.42
1.53
0.805


10
0.00
3.20
8.17
10.7
9.08
10.7
11.8
11.2
9.08
6.20
3.38
2.75
1.84
0.672


11
0.00
4.20
6.86
6.36
9.97
11.3
11.3
10.2
7.79
5.08
4.38
2.67
1.53
0.815


12
0.00
4.73
7.71
9.48
11.9
15.1
16.5
15.5
13.2
8.89
4.58
3.60
2.67
2.12


13
0.00
1.56
2.87
3.89
6.31
7.43
7.87
7.64
7.01
5.34
3.57
2.12
1.35
1.41


14
0.00
0.663
2.20
3.86
8.74
14.7
15.0
15.3
13.6
10.7
6.84
4.47
2.39
1.59


MEAN
0
3.02
5.76
7.46
10.1
12
12.4
11.8
9.42
7.18
5.08
3.58
2.24
1.22


SD
0
1.53
2.45
2.53
2.03
2.45
2.61
2.81
2.77
2.27
1.48
0.943
0.556
0.408


% CV
0
50.7
42.5
33.9
20.1
20.4
21
23.8
29.4
31.6
29.1
26.3
24.8
33.4
















TABLE 17







Hydrocodone Plasma Concentration (ng/mL) after administration of two (2)


Immediate-Release Hydrocodone 7.5 mg/Acetaminophen 500 mg tablets-Formulation C









Time (hours)





















Subject
−0.08
0.5
0.75
1
2
3
4
6
9
12
18
24
30
36
























1
0.00
40.6
41.6
45.4
32.1
26.3
22.7
15.2
9.95
6.08
2.58
1.20
0.585
0.00


2
0.00
44.3
50.7
40.1
28.6
23.3
20.2
15.6
9.46
6.08
2.96
1.68
0.872
0.00


3
0.00
17.6
42.3
42.6
37.8
35.4
31.2
21.0
13.0
7.79
3.12
1.77
0.685
0.00


4
0.00
21.2
43.3
36.5
26.9
23.5
20.7
15.4
9.39
5.09
2.27
1.17
0.523
0.00


5
0.00
37.4
39.3
36.1
27.9
22.4
18.1
14.1
7.91
4.98
2.37
1.07
0.546
0.00


6
0.00
3.17
8.67
16.3
17.5
16.9
13.8
11.3
6.52
4.22
1.71
0.703
0.00
0.00


7
0.00
0.900
6.76
14.7
18.3
17.1
14.1
9.66
5.52
3.32
1.21
0.00
0.00
0.00


8
0.00
2.97
13.7
22.2
32.4
28.8
24.2
18.3
10.9
6.46
2.17
1.02
0.00
0.00


9
0.00
50.0
39.3
33.7
24.2
20.1
17.0
13.0
6.84
4.01
1.47
0.565
0.00
0.00


10
0.00
0.627
14.8
25.2
22.4
17.3
16.5
10.9
5.90
3.15
1.05
0.00
0.00
0.00


11
0.00
8.46
13.3
29.3
31.3
24.8
21.0
14.0
9.43
6.04
2.62
1.14
0.00
0.00


12
0.00
30.6
44.4
44.4
40.0
30.8
29.1
19.9
11.3
6.86
3.15
1.47
0.634
0.00


13
0.00
3.73
12.2
17.9
19.1
19.8
16.3
13.9
8.72
5.43
2.51
0.706
0.00
0.00


14
0.00
18.0
29.7
35.3
30.7
26.6
23.4
16.1
9.20
6.24
2.60
1.27
0.556
0.00


MEAN
0.00
20.0
28.6
31.4
27.8
23.8
20.6
14.9
8.86
5.41
2.27
0.983
0.314
0.00


SD
0.00
17.7
16.0
10.6
6.93
5.48
5.21
3.26
2.15
1.36
0.676
0.541
0.336
0.00


% CV
0.00
88.5
55.9
33.8
24.9
23.0
25.3
21.9
24.3
25.1
29.8
55.0
107
0.00
















TABLE 18







Hydrocodone Plasma Concentration (ng/mL) after administration of one (1)


Controlled-Release Hydrocodone Bitartrate 15 mg capsule-Formulation D









Time (hours)





















Subject
−0.08
0.5
0.75
1
2
3
4
6
9
12
18
24
30
36
























1
0.00
1.76
4.07
5.17
8.33
9.72
11.1
14.0
13.6
11.7
8.78
6.14
3.91
1.97


2
0.00
2.76
4.83
5.13
6.17
10.4
10.6
13.5
11.8
10.1
6.57
3.71
2.57
1.34


3
0.00
2.91
4.25
6.01
10.1
12.3
12.0
14.8
13.5
11.4
7.40
4.16
2.65
1.46


4
0.00
1.69
5.93
6.26
8.29
8.37
8.06
10.5
8.91
8.70
4.58
2.61
1.63
0.536


5
0.00
0.616
2.74
4.47
8.58
9.16
8.60
10.1
8.66
6.64
4.72
2.57
2.05
0.986


6
0.00
0.663
2.40
4.87
7.50
10.1
11.7
13.0
11.5
8.30
5.38
3.88
2.39
1.25


7
0.00
0.00
1.55
2.32
4.61
6.38
7.22
7.41
6.75
4.82
3.10
1.72
0.984
0.578


8
0.00
1.26
3.03
5.15
7.26
8.80
8.81
9.34
9.07
9.28
6.81
3.31
1.93
1.25


9
0.00
3.36
3.63
6.38
8.31
8.04
8.20
9.55
8.28
6.49
3.72
2.25
1.92
0.901


10
0.00
0.692
2.91
2.95
5.11
6.09
7.37
7.11
6.33
5.67
3.76
2.76
1.43
0.573


11
0.00
1.11
2.87
3.28
6.82
9.69
10.3
12.0
12.2
8.81
5.76
3.25
2.10
1.08


12
0.00
2.25
3.31
4.72
8.03
11.4
11.2
12.1
11.0
9.75
5.64
3.51
2.71
1.34


13
0.00
0.00
1.29
2.71
5.51
6.67
8.92
8.44
7.13
7.01
3.99
2.41
1.04
0.858


14
0.00
1.02
2.94
4.53
8.82
10.5
11.7
14.1
13.0
10.2
6.37
3.56
1.93
1.61


MEAN
0.00
1.44
3.27
4.57
7.39
9.12
9.70
11.1
10.1
8.49
5.47
3.27
2.09
1.12


SD
0.00
1.06
1.23
1.31
1.57
1.86
1.71
2.57
2.55
2.11
1.61
1.08
0.754
0.419


% CV
0.00
73.6
37.6
28.7
21.2
20.4
17.6
23.2
25.2
24.9
29.4
33.0
36.1
37.4









The pharmacokinetic parameters are set forth in Table 19 below:












TABLE 19





Parameter
Meana
% Ratiob,c
90% Clb





















Ex. 1
IR Ex.
Ex. 1/IR Ex.





Fasted
Fasted
Fasted
Lower
Upper





AUC(0, last)
200.95
216.35
93.36
86.96
100.23


(ng · hr/mL)







Cmax (ng/mL)
13.16
33.37
39.48
35.26
44.20


Tmax (hr)
4.07
1.32
208.11
257.17
357.80


W50 (hr)
13.41
4.67
287.38
265.91
314.15


T½(abs) (hr)
1.64
0.69
237.65
197.73
284.44


T½(elim) (hr)
6.44
3.09
208.78
184.43
234.20






Ex. 2
IR Ex.
Ex. 2/IR Ex.





Fasted
Fasted
Fasted
Lower
Upper





AUC(0, last)
201.57
216.35
93.21
86.82
100.07


(ng · hr/mL)







Cmax (ng/mL)
12.42
33.37
37.36
33.37
41.83


Tmax (hr)
4.20
1.32
317.57
262.19
362.83


W50 (hr)
13.08
4.67
280.31
257.03
305.26


T½(abs) (hr)
1.57
0.69
227.91
183.84
270.55


T½(elim) (hr)
7.86
3.09
254.85
231.54
281.31






Ex. 3
IR. Ex.
Ex. 3/IR Ex.





Fasted
Fasted
Fasted
Lower
Upper





AUC(0, last)
194.40
216.35
90.28
84.09
96.92


(ng · hr/mL)







Cmax (ng/mL)
10.93
33.37
32.69
29.20
36.60


Tmax (hr)
5.93
1.32
448.65
398.87
499.51


W50 (hr)
16.30
4.67
349.21
328.68
376.92


T½(abs) (hr)
2.98
0.69
431.26
395.95
482.67


T½(elim) (hr)
6.96
3.09
225.61
200.49
250.26






aGeometric means for AUC(0, last) and Cmax and arithmetic means for Tmax, W50, T½(abs), and T½(elim).




bRatio and 90% Cl are based on least square means.




cRatio (%): (Test mean/Reference mean) × 100, based on least square means







Example 8

Hydrocodone sustained release tablets were produced with the formula set forth in Table XX below:












TABLE XX






Ingredient
mg/tab
kg/batch



















Hydrocodone bitartrate
15
15.0



Dibasic calcium phosphate
31
31.0



Glyceryl behenate
10
10.0



Stearyl alcohol
22
22.0



Microcrystalline cellulose
31
31.0



Magnesium stearate
1.0
1.0



Opadry Purple YS-1-10371-A
5.0
5.0



Purified water
N/A1
28.331




115.0 mg
115.0 kg






1Evaporates during processing and is not part of finished product.








According to the following procedure:
  • 1. Milling: Pass stearyl alcohol flakes through a mill.
  • 2. Blending: Mix The Hydrocodone bitartrate, Dibasic calcium phosphate, Glyceryl behenate, Stearyl alcohol and Microcrystalline cellulose with a suitable blender
  • 3. Extrusion: Continuously feed the blended material into a twin screw extruder at an elevated temperature to soften and form an extrudate.
  • 4. Cooling: Allow the extrudate to cool on a Conveyor.
  • 5. Milling: Pass the cooled extrudate through a mill to obtain a suitable particle size granulation
  • 6. Blending: Blend the milled extrudate with the magnesium stearate.
  • 7. Compression: Compress the resultant granulation using a tablet press.
  • 8. Coating: Prepare a film coating solution by dispersing the Opadry in Purified Water and applying it to the tablet cores.


The tablets were then tested for dissolution using the following procedure:

  • 1. Apparatus: USP Type I (basket), 100 rpm.
  • 2. Medium: 700 ml SGF (without enzymes) for first 55 minutes, thereafter made 900 ml with phosphate buffer to pH 7.5.
  • 3. Sampling time: 1, 2, 4, 8, and 12 hours.
  • 4. Analytical: High Performance Liquid Chromatography.


The dissolution parameters are set forth in Table XXI below:











TABLE XXI






Time (hrs)
% Dissolved


















1
22



2
37



4
58



8
84



12
99









Example 9

A 3 way crossover, pharmacokinetic comparison study of a single dose of 15 mg Hydrocodone Controlled Release Tablets (Example 8) in Fed and Fasted and of 15 mg Hydrocodone Immediate Release (2×7.5 mg tablets) was given over two Q6H doses in fasted normal volunteers.


The Cmax and Tmax were then obtained for Example 8 and an immediate release reference standard in a bioavailability study, as set forth in Table XXII and XXIII below:











TABLE XXII






Pharmacokinetic data




(Fasted State)
Hydrocodone Bitartrate


















Cmax (ng/ml)
43.16



IR reference product




(Dose adjusted)




Cmax (ng/ml)
17.87



CR product




Cmax (CR)/Cmax
41%



(IR)




Tmax (hr)
6.42



IR reference product




Tmax (hr)
4.04



CR product



















TABLE XXXIII








Hydrocodone



Hydrocodone
Hydrocodone
Bitartrate IR



Bitartrate
Bitartrate
2 × 7.5 mg


Pharmacokinetic
CR 15 mg Tablets
CR 15 mg Tablets
Tablets


data
(Fasted)
(Fed)
(Fasted)


















Cmax (ng/ml)
17.87
19.23
21.58


C12 hour
11.06
12.84



C12 hour/Cmax
62%
67%



Tmax (hr)
4.04
4.81
6.42


AUC
267.43
277.58
229.33








Claims
  • 1. A process for the preparation of a solid oral dosage form comprising: compressing a therapeutically effective amount of hydrocodone and one or more excipients into a tablet such that the tablet releases hydrocodone at such a rate that plasma concentrations of hydrocodone are maintained within the therapeutic range but below toxic concentrations for about 12 hours or longer, andprovides a hydrocodone plasma concentration of at least about 8 ng/ml at about 8 hours after administration, anda hydrocodone plasma concentration of at least 5 ng/ml at 12 hours after administration.
  • 2. The process of claim 1, wherein the dosage form provides controlled release of hydrocodone for about 12 hours.
  • 3. The process of claim 1, wherein the dosage form provides controlled release of hydrocodone for about 24 hours.
  • 4. The process of claim 3, wherein the dosage form provides a hydrocodone plasma concentration of at least about 8 ng/ml at about 2 hours after administration.
  • 5. The process of claim of claim 4, wherein one of the excipients is a pharmaceutically acceptable polymer.
  • 6. The process of claim 5, wherein the pharmaceutically acceptable polymer comprises between 1% and 80% of the tablet by weight.
  • 7. The process of claim 1, wherein one of the excipients is a material having a melting point of from 30 to about 200° C.
  • 8. The process of claim 5, wherein the pharmaceutically acceptable polymer is a hydroxyalkylcellulose.
  • 9. The process of claim 8, wherein the hydroxyalkylcellulose is a hydroxypropylcellulose or a hydroxypropylmethylcellulose.
  • 10. The process of claim 9, wherein the hydroxyalkylcellulose is the hydroxypropylmethylcellulose.
  • 11. The process of claim 7, wherein the material is a polyalkylene glycol.
  • 12. The process of claim 1, wherein the dosage form provides a Cmax that increases linearly from one dosage strength to another.
  • 13. The process of claim 3, wherein the dosage form provides a plasma concentration profile of hydrocodone with a C12/Cmax hydrocodone ratio of 0.55 to 0.85.
  • 14. The process of claim 3, wherein the dosage form provides a plasma concentration profile of hydrocodone with a C12/Cmax hydrocodone ratio of 0.55 to 0.65.
  • 15. The process of claim 3, wherein the dosage form provides a plasma concentration profile of hydrocodone with a C12/Cmax hydrocodone ratio of 0.65 to 0.85.
  • 16. The process of claim 14, wherein said C12/Cmax hydrocodone ratio is calculated from hydrocodone plasma concentrations from first administration of the dosage form to a population of fasted humans.
  • 17. The process of claim 15, wherein said C12/Cmax hydrocodone ratio is calculated from hydrocodone plasma concentrations from first administration of the dosage form to a population of fasted humans.
  • 18. The process of claim 14, wherein a portion of the plasma concentration profile of hydrocodone is relatively flat.
  • 19. The process of claim 14, wherein said C12/Cmax hydrocodone ratio is calculated from hydrocodone plasma concentrations from first administration of the dosage form to a fed human.
  • 20. The process of claim 15, wherein said C12/Cmax hydrocodone ratio is calculated from hydrocodone plasma concentrations from first administration of the dosage form to a fed human.
  • 21. The process of claim 13, wherein said C12/Cmax hydrocodone ratio is calculated from hydrocodone plasma concentrations from first administration of the dosage form to a population of fed humans.
  • 22. The process of claim 1, wherein said hydrocodone plasma concentrations are hydrocodone plasma concentrations from first administration of the dosage form to a fasted human.
  • 23. The process of claim 1, wherein said hydrocodone plasma concentrations are hydrocodone plasma concentrations from first administration of the dosage form to a fed human.
  • 24. A process for the preparation of a solid oral dosage form comprising: formulating a tablet comprising from about 5 mg to about 1250 mg of hydrocodone and one or more excipients such that the tablet providesa controlled release of hydrocodone over about 8 to about 24 hours,a hydrocodone plasma concentration of at least about 8 ng/ml at about 8 hours after administration, anda hydrocodone plasma concentration of at least 5 ng/ml at 12 hours after administration.
  • 25. The process of claim 24, wherein the tablet comprises a bitartrate salt of hydrocodone.
  • 26. The process of claim 24, wherein the dosage form provides a hydrocodone plasma concentration of at least about 8 ng/ml at about 2 hours after administration.
  • 27. The process of claim 24, wherein the dosage form provides a plasma concentration profile of hydrocodone with a C12/Cmax hydrocodone ratio of 0.55 to 0.85.
  • 28. The process of claim 27, wherein a portion of the plasma concentration profile of hydrocodone is relatively flat.
  • 29. The process of claim 27, wherein said C12/Cmax hydrocodone ratio is calculated from hydrocodone plasma concentrations from first administration of the dosage form to a fasted human.
  • 30. The process of claim 24, wherein said hydrocodone plasma concentrations are hydrocodone plasma concentrations from first administration of the dosage form to a fed human.
  • 31. A process for the preparation of a solid oral dosage form comprising: compressing a therapeutically effective amount of hydrocodone and one or more excipients into a tablet such that the tabletreleases hydrocodone at such a rate that plasma concentrations of hydrocodone are maintained within the therapeutic range but below toxic concentrations for about 12 hours or longer andprovides a hydrocodone plasma concentration of at least 6 ng/ml at about 12 hours after first administration, a hydrocodone plasma concentration of at least about 8 ng/ml at about 8 hours after administration, and a C12/Cmax hydrocodone ratio of 0.55 to 0.85.
  • 32. A process for the preparation of a solid oral dosage form comprising: formulating a tablet comprising from about 5 mg to about 1250 mg of hydrocodone and one or more excipients such that the tablet providesa controlled release of hydrocodone over about 8 to about 24 hours,a hydrocodone plasma concentration of at least 6 ng/ml at about 12 hours after first administration,a hydrocodone plasma concentration of at least about 8 ng/ml at about 8 hours after administration, anda C12/Cmax hydrocodone ratio of 0.55 to 0.85.
Parent Case Info

This application is a continuation of U.S. Ser. No. 15/134,901, filed on Apr. 21, 2016, which is a continuation of U.S. Ser. No. 14/672,894, filed on Mar. 30, 2015, now U.S. Pat. No. 9,320,717, which is a continuation of U.S. Ser. No. 14/635,198, filed on Mar. 2, 2015, now U.S. Pat. No. 9,056,107, which is a continuation of U.S. Ser. No. 14/094,968, filed on Dec. 3, 2013, which is a continuation of U.S. Ser. No. 13/833,263, filed on Mar. 15, 2013, which is a continuation of U.S. Ser. No. 12/982,386, filed on Dec. 30, 2010, now U.S. Pat. No. 9,980,291, which is a continuation of U.S. Ser. No. 10/864,829, filed on Jun. 9, 2004, now U.S. Pat. No. 7,943,174, which is a continuation of U.S. Ser. No. 09/702,283, filed on Oct. 30, 2000, which claims priority from U.S. Provisional Application Ser. No. 60/162,541 filed Oct. 29, 1999, all hereby incorporated by reference.

US Referenced Citations (367)
Number Name Date Kind
2738303 Blythe Mar 1956 A
3424839 Montandraud Jan 1969 A
3634584 Poole Jan 1972 A
3845770 Theeuwes et al. Nov 1974 A
3870790 Lowey et al. Mar 1975 A
3916889 Russell Nov 1975 A
3916899 Theeuwes et al. Nov 1975 A
4063064 Saunders et al. Dec 1977 A
4088864 Theeuwes et al. May 1978 A
4132753 Blichare et al. Jan 1979 A
4377568 Chopra Mar 1983 A
4385078 Onda et al. May 1983 A
4389393 Schor et al. Jun 1983 A
4421736 Walters Dec 1983 A
4443428 Oshlack Apr 1984 A
4464378 Hussain Aug 1984 A
4483847 Augart Nov 1984 A
4520172 Lehmann et al. May 1985 A
4539199 Orban et al. Sep 1985 A
4548990 Mueller et al. Oct 1985 A
4557925 Lindahl et al. Dec 1985 A
4600645 Ghebre-Sellassie et al. Jul 1986 A
4609542 Panoz et al. Sep 1986 A
4612008 Wong et al. Sep 1986 A
4708874 DeHaan et al. Nov 1987 A
4728512 Mehta et al. Mar 1988 A
4728513 Ventouras Mar 1988 A
4794001 Mehta et al. Dec 1988 A
4797410 El-Fakahany Jan 1989 A
4806337 Snipes et al. Feb 1989 A
4814176 Makino et al. Mar 1989 A
4828836 Elger et al. May 1989 A
4834984 Goldie et al. May 1989 A
4834985 Elger et al. May 1989 A
4844896 Bohm et al. Jul 1989 A
4844909 Goldie et al. Jul 1989 A
4861596 Curtis et al. Aug 1989 A
4861598 Oshlack Aug 1989 A
4863456 Stephens et al. Sep 1989 A
4873092 Azuma et al. Oct 1989 A
4888178 Rotini et al. Dec 1989 A
4892742 Shah Jan 1990 A
4894234 Sharma et al. Jan 1990 A
4904476 Mehta et al. Feb 1990 A
4935246 Ahrens Jun 1990 A
4940588 Sparks et al. Jul 1990 A
4948586 Bohm et al. Aug 1990 A
4952402 Sparks et al. Aug 1990 A
4956182 Bequette et al. Sep 1990 A
4959219 Chow et al. Sep 1990 A
4970075 Oshlack Nov 1990 A
4971805 Kitanishi et al. Nov 1990 A
4983730 Domeshek et al. Jan 1991 A
4990341 Goldie et al. Feb 1991 A
4996047 Kelleher et al. Feb 1991 A
5002774 Agrawala et al. Mar 1991 A
5007790 Shell Apr 1991 A
5019397 Wong et al. May 1991 A
5023088 Wong et al. Jun 1991 A
5023089 Sakamoto et al. Jun 1991 A
5024842 Edgren et al. Jun 1991 A
5026560 Makino et al. Jun 1991 A
5030400 Danielsen et al. Jul 1991 A
5068110 Fawzi et al. Nov 1991 A
5071646 Malkowska et al. Dec 1991 A
5102668 Eichel et al. Apr 1992 A
5122384 Paradissis et al. Jun 1992 A
5126145 Evenstad et al. Jun 1992 A
5132142 Jones et al. Jul 1992 A
5133974 Paraqdissis et al. Jul 1992 A
5158777 Abramowitz et al. Oct 1992 A
5162117 Stupak et al. Nov 1992 A
5167964 Muhammad et al. Dec 1992 A
5169645 Shukla et al. Dec 1992 A
5178868 Malmqvist et al. Jan 1993 A
5178878 Wehling et al. Jan 1993 A
5196203 Boehm Mar 1993 A
5202128 Morella et al. Apr 1993 A
5206030 Wheatley et al. Apr 1993 A
5219575 Von Bommel et al. Jun 1993 A
5226902 Bae et al. Jul 1993 A
5229131 Amidon et al. Jul 1993 A
5248516 Wheatley et al. Sep 1993 A
5258436 Wheatley et al. Nov 1993 A
5260068 Chen Nov 1993 A
5260069 Chen Nov 1993 A
5262173 Sheth et al. Nov 1993 A
5266331 Oshlack et al. Nov 1993 A
5273758 Royce Dec 1993 A
5273760 Oshlack et al. Dec 1993 A
5283065 Doyon et al. Feb 1994 A
5286493 Oshlack et al. Feb 1994 A
5292461 Juch et al. Mar 1994 A
5320853 Noda et al. Jun 1994 A
5321012 Mayer et al. Jun 1994 A
5330759 Pagay et al. Jul 1994 A
5330766 Morella Jul 1994 A
5352683 Mayer et al. Oct 1994 A
5354556 Sparks et al. Oct 1994 A
5356467 Oshlack et al. Oct 1994 A
5378474 Morella et al. Jan 1995 A
5380790 Chen et al. Jan 1995 A
5384130 Kamada Jan 1995 A
5387421 Amidon et al. Feb 1995 A
5395628 Noda et al. Mar 1995 A
5401512 Rhodes et al. Mar 1995 A
5403593 Royce Apr 1995 A
5411745 Oshlack et al. May 1995 A
5436011 Dennis et al. Jul 1995 A
5439689 Hendrickson et al. Aug 1995 A
5445828 Pozzi et al. Aug 1995 A
5445829 Parasissis et al. Aug 1995 A
5456923 Nakamichi et al. Oct 1995 A
5458879 Singh et al. Oct 1995 A
5460817 Langley et al. Oct 1995 A
5460826 Merrill et al. Oct 1995 A
5460828 Santus et al. Oct 1995 A
5472708 Chen Dec 1995 A
5472712 Oshlack et al. Dec 1995 A
5478577 Sacler et al. Dec 1995 A
5484608 Rudmie et al. Jan 1996 A
5490990 Grabowski et al. Feb 1996 A
5500227 Oshlack et al. Mar 1996 A
5502058 Mayer et al. Mar 1996 A
RE35200 Lehmann et al. Apr 1996 E
5503846 Wehling et al. Apr 1996 A
5508040 Chen Apr 1996 A
5508042 Oshlack et al. Apr 1996 A
5518730 Fuisz May 1996 A
5520931 Persson et al. May 1996 A
5534263 Wong et al. Jul 1996 A
5549912 Oshlack et al. Aug 1996 A
5580578 Oshlack et al. Dec 1996 A
5593694 Hayashida et al. Jan 1997 A
5593695 Merrill et al. Jan 1997 A
5601842 Bartholomaeus Feb 1997 A
5614218 Olsson et al. Mar 1997 A
5629011 Illum May 1997 A
5629017 Pozzi et al. May 1997 A
5637320 Bourke et al. Jun 1997 A
5639476 Oshlack et al. Jun 1997 A
5654006 Femandez et al. Aug 1997 A
5656295 Oshlack et al. Aug 1997 A
5667805 Merrill et al. Sep 1997 A
5670172 Buxton et al. Sep 1997 A
5672360 Sackler et al. Sep 1997 A
5681584 Savastano et al. Oct 1997 A
5681585 Oshlack et al. Oct 1997 A
5726316 Crooks et al. Mar 1998 A
5731006 Akiyama et al. Mar 1998 A
5744166 Illum et al. Apr 1998 A
5753261 Femandez et al. May 1998 A
5776856 Narayanan Jul 1998 A
5783212 Fassihi et al. Jul 1998 A
5807579 Vikov et al. Sep 1998 A
5811126 Krishnamurthy et al. Sep 1998 A
5820879 Fernandez et al. Oct 1998 A
5820883 Tice et al. Oct 1998 A
5834023 Chen Nov 1998 A
5834024 Lepore Nov 1998 A
5837284 Mehta et al. Nov 1998 A
5840329 Bai Nov 1998 A
5840332 Lerner et al. Nov 1998 A
5840754 Guittard et al. Nov 1998 A
5843477 Alexander Dec 1998 A
5843480 Miller et al. Dec 1998 A
5849240 Miller et al. Dec 1998 A
5851555 Sanghvi et al. Dec 1998 A
5858412 Staniforth et al. Jan 1999 A
5866161 Chiders et al. Feb 1999 A
5866164 Kuczynski et al. Feb 1999 A
5874090 Baker et al. Feb 1999 A
5879705 Heafield et al. Mar 1999 A
5885616 Hsiao et al. Mar 1999 A
5891471 Miller et al. Apr 1999 A
5904927 Amiji May 1999 A
5948787 Merrill Sep 1999 A
5955104 Momberger et al. Sep 1999 A
5958452 Oshlack et al. Sep 1999 A
5958458 Norling et al. Sep 1999 A
5958459 Chasin et al. Sep 1999 A
5958551 Oshlack Oct 1999 A
5965161 Oshlack et al. Oct 1999 A
5965163 Miller et al. Oct 1999 A
5968551 Oshlack et al. Oct 1999 A
5968661 Saito et al. Oct 1999 A
6024981 Khankari et al. Feb 2000 A
6025502 Winkler et al. Feb 2000 A
6039980 Baichwal Mar 2000 A
6096148 Kingma Aug 2000 A
6103219 Sherwood et al. Aug 2000 A
6103261 Chasin et al. Aug 2000 A
6114423 Eck et al. Sep 2000 A
6129933 Oshlack et al. Oct 2000 A
6143322 Sackler et al. Nov 2000 A
6143353 Oshlack et al. Nov 2000 A
6155423 Katzner et al. Dec 2000 A
6156342 Sriwongjanya et al. Dec 2000 A
6159501 Skinhoj et al. Dec 2000 A
6162467 Miller et al. Dec 2000 A
6194005 Farah et al. Feb 2001 B1
6200604 Pather et al. Mar 2001 B1
6217904 Midha et al. Apr 2001 B1
6228398 Devane et al. May 2001 B1
6238704 Suzuki et al. May 2001 B1
6245351 Nara et al. Jun 2001 B1
6245357 Edgren et al. Jun 2001 B1
6251430 Zhang et al. Jun 2001 B1
6261599 Oshlack et al. Jul 2001 B1
6262072 Lee Jul 2001 B1
6264983 Upadhyay et al. Jul 2001 B1
6290990 Grabowski et al. Sep 2001 B1
6294195 Oshlack et al. Sep 2001 B1
6294591 Blum et al. Sep 2001 B1
6300403 Mayer et al. Oct 2001 B1
6309668 Bastin et al. Oct 2001 B1
6316031 Oshlack et al. Nov 2001 B1
6322819 Burnside et al. Nov 2001 B1
6335033 Oshlack et al. Jan 2002 B2
6340476 Midha et al. Jan 2002 B1
6344215 Bettman et al. Feb 2002 B1
6368625 Siebert et al. Apr 2002 B1
6372254 Ting et al. Apr 2002 B1
6375987 Farah et al. Apr 2002 B1
6387404 Oshlack et al. May 2002 B2
6399096 Miller et al. Jun 2002 B1
6419954 Chu et al. Jul 2002 B1
6419960 Krishnamurthy et al. Jul 2002 B1
6488962 Berner Dec 2002 B1
6491945 Childers et al. Dec 2002 B1
6500459 Chhabra et al. Dec 2002 B1
6534091 Garces et al. Mar 2003 B1
6552031 Burch et al. Apr 2003 B1
6562375 Sako et al. May 2003 B1
6572885 Oshlack et al. Jun 2003 B2
6589960 Harclerode et al. Jul 2003 B2
6599529 Skinhoj et al. Jul 2003 B1
6607750 Upadhyay et al. Aug 2003 B2
6680071 Johnson et al. Jan 2004 B1
6685964 Bartholomaus et al. Feb 2004 B1
6696066 Kaiko et al. Feb 2004 B2
6699502 Fanara et al. Mar 2004 B1
6706281 Oshlack et al. Mar 2004 B2
6730321 Ting et al. May 2004 B2
6730325 Devane et al. May 2004 B2
6733783 Oshlack et al. May 2004 B2
6733790 Garces et al. May 2004 B1
6743442 Oshlack et al. Jun 2004 B2
6753014 Sjoblom et al. Jun 2004 B1
6759059 Pettersson et al. Jul 2004 B1
6780504 Rupprecht et al. Aug 2004 B2
6793936 Devane et al. Sep 2004 B2
6863901 Hirsh et al. Mar 2005 B2
6902742 Devane et al. Jun 2005 B2
6905709 Oshlack et al. Jun 2005 B2
7022313 O'Connor et al. Apr 2006 B2
7090867 Odidi et al. Aug 2006 B2
7387792 Hirsh et al. Jun 2008 B2
7399488 Hirsh et al. Jul 2008 B2
7442387 Sugihara et al. Oct 2008 B2
7514100 Oshlack et al. Apr 2009 B2
7658939 Oshlack et al. Feb 2010 B2
7776314 Bartholomaus et al. Aug 2010 B2
7790215 Sackler et al. Sep 2010 B2
7846476 Oshlack et al. Dec 2010 B2
7943174 Oshlack et al. May 2011 B2
8142811 Oshlack et al. Mar 2012 B2
8231898 Oshlack et al. Jul 2012 B2
8361499 Oshlack et al. Jan 2013 B2
8551520 Oshlack et al. Oct 2013 B2
8647667 Oshlack et al. Feb 2014 B2
8715721 Oshlack et al. May 2014 B2
8951555 Oshlack et al. Feb 2015 B1
8980291 Oshlack et al. Mar 2015 B2
9023401 Oshlack et al. May 2015 B1
9056052 Oshlack et al. Jun 2015 B1
9056107 Oshlack et al. Jun 2015 B1
9060940 Oshlack et al. Jun 2015 B2
9198863 Oshlack et al. Dec 2015 B2
9205055 Oshlack et al. Dec 2015 B2
9205056 Oshlack et al. Dec 2015 B2
9289391 Oshlack et al. Mar 2016 B2
20020012675 Jain et al. Jan 2002 A1
20020044966 Bartholomaus et al. Apr 2002 A1
20020110595 Chang et al. Aug 2002 A1
20020110598 Chung et al. Aug 2002 A1
20030077297 Chen et al. Apr 2003 A1
20030091630 Louie-Helm May 2003 A1
20030180362 Park et al. Sep 2003 A1
20030190358 Oshlack et al. Oct 2003 A1
20040009219 Odidi et al. Jan 2004 A1
20040028735 Kositprapa Feb 2004 A1
20040052844 Hsiao et al. Mar 2004 A1
20040096499 Vaya et al. May 2004 A1
20040096500 Oshlack et al. May 2004 A1
20040105887 Oshlack et al. Jun 2004 A1
20040121001 Oshlack et al. Jun 2004 A1
20040131552 Boehm Jul 2004 A1
20040142035 Chang et al. Jul 2004 A1
20040151791 Mayo-Alvarez et al. Aug 2004 A1
20040157784 Chopdekar et al. Aug 2004 A1
20040170680 Oshlack et al. Sep 2004 A1
20040185098 Oshlack et al. Sep 2004 A1
20040197405 Devane et al. Oct 2004 A1
20040208930 Yoneda et al. Oct 2004 A1
20040208936 Chorin et al. Oct 2004 A1
20040224017 Mulye Nov 2004 A1
20040253310 Fischer et al. Dec 2004 A1
20040266807 Oshlack et al. Dec 2004 A1
20050020613 Boehm et al. Jan 2005 A1
20050053656 Ping Mar 2005 A1
20050074493 Mehta et al. Apr 2005 A1
20050089568 Oshlack et al. Apr 2005 A1
20050106249 Hwang et al. May 2005 A1
20050163856 Maloney et al. Jul 2005 A1
20050165038 Gordon Jul 2005 A1
20050169989 Moe et al. Aug 2005 A1
20050169990 Kao et al. Aug 2005 A1
20050245483 Brogmann et al. Nov 2005 A1
20050266072 Oshlack et al. Dec 2005 A1
20060003004 Hirsh et al. Jan 2006 A1
20060024361 Odidi et al. Feb 2006 A1
20060104909 Vaghefi et al. May 2006 A1
20060153915 Park et al. Jul 2006 A1
20060153916 Vaya et al. Jul 2006 A1
20060204573 Mulye Sep 2006 A1
20060233879 Lerner et al. Oct 2006 A1
20060233880 Lerner et al. Oct 2006 A1
20060240105 Devane Oct 2006 A1
20060251721 Cruz et al. Nov 2006 A1
20060263429 Feng Nov 2006 A1
20060269604 Sackler et al. Nov 2006 A1
20070003617 Fischer et al. Jan 2007 A1
20070009598 Marechal et al. Jan 2007 A1
20070020335 Chen et al. Jan 2007 A1
20070203165 Shafer et al. Aug 2007 A1
20070212414 Baichwal et al. Sep 2007 A1
20070231268 Emigh et al. Oct 2007 A1
20070281021 McKinney et al. Dec 2007 A1
20080031901 Qiu et al. Feb 2008 A1
20080069891 Wabib Mar 2008 A1
20080102121 Devane et al. May 2008 A1
20080113025 Devane et al. May 2008 A1
20080132532 Wright et al. Jun 2008 A1
20080226734 Jenkins et al. Sep 2008 A1
20080311205 Habib et al. Dec 2008 A1
20090149479 Jenkins et al. Jun 2009 A1
20090238868 Mehta Sep 2009 A1
20090297617 Rariy et al. Dec 2009 A1
20090304793 Boehm Dec 2009 A1
20090317355 Roth et al. Dec 2009 A1
20100010030 Jain et al. Jan 2010 A1
20100015223 Cailly-Dufestel et al. Jan 2010 A1
20100040689 Hou Feb 2010 A1
20100098771 Mehta Apr 2010 A1
20100172989 Roth et al. Jul 2010 A1
20100249045 Babul Sep 2010 A1
20100323016 Nadjsombati Dec 2010 A1
20110065628 Johnson et al. Mar 2011 A1
20110262532 Oshlack et al. Oct 2011 A1
20120164220 Huang Jun 2012 A1
20120165358 Cruz et al. Jun 2012 A1
20130209560 Hamed et al. Aug 2013 A1
20130295177 Oshlack et al. Nov 2013 A1
20130302418 Oshlack et al. Nov 2013 A1
20140112981 Oshlack et al. Apr 2014 A1
20140161879 Devane et al. Jun 2014 A1
Foreign Referenced Citations (127)
Number Date Country
9047732 Jul 1990 AU
9341654 Feb 1995 AU
1999 44493 Oct 1999 AU
2082573 Nov 1992 CA
2131350 Jan 1994 CA
0097523 Jun 1983 EP
0097523 Jan 1984 EP
0108218 May 1984 EP
0147780 Jul 1985 EP
0202051 Nov 1986 EP
0235986 Feb 1987 EP
0253104 Jan 1988 EP
0267702 May 1988 EP
0271193 Jun 1988 EP
0274734 Jul 1988 EP
0311582 Apr 1989 EP
0327295 Aug 1989 EP
0377518 Jan 1990 EP
0388954 Mar 1990 EP
0361910 Apr 1990 EP
0377517 Jul 1990 EP
0377518 Jul 1990 EP
0388954 Sep 1990 EP
0415693 Mar 1991 EP
0452145 Oct 1991 EP
0502642 Feb 1992 EP
0534628 Apr 1992 EP
0499299 Aug 1992 EP
0532348 Sep 1992 EP
0533297 Mar 1993 EP
0535841 Apr 1993 EP
0546676 Jun 1993 EP
0548448 Jun 1993 EP
0553392 Aug 1993 EP
0 338 444 Nov 1993 EP
0580860 Feb 1994 EP
0636370 Jun 1994 EP
0361680 Jul 1994 EP
0609961 Aug 1994 EP
0430287 Oct 1994 EP
0636370 Feb 1995 EP
0647448 Apr 1995 EP
0665010 Aug 1995 EP
0965343 Dec 1999 EP
1419766 May 2004 EP
1442745 Aug 2004 EP
1504757 Feb 2005 EP
1782834 May 2007 EP
2053681 Apr 1984 GB
2170104 Jul 1986 GB
2178313 Feb 1987 GB
2179254 Mar 1997 GB
202403 Apr 1998 HU
40081086 Apr 1992 JP
08157392 Jun 1996 JP
2 069 558 Nov 1996 RU
WO 9201446 Feb 1992 WO
WO 9202209 Feb 1992 WO
WO 9204011 Mar 1992 WO
WO 9206679 Apr 1992 WO
WO 9208459 May 1992 WO
WO 9304675 Mar 1993 WO
WO 9307859 Apr 1993 WO
WO 9307861 Apr 1993 WO
WO 9310765 Jun 1993 WO
WO 9318753 Sep 1993 WO
WO 9403160 Feb 1994 WO
WO 9403161 Feb 1994 WO
WO 9405262 Mar 1994 WO
WO 9422431 Oct 1994 WO
W0-9428882 Dec 1994 WO
WO 9514460 Jun 1995 WO
WO 9600066 Jan 1996 WO
WO 9601629 Jan 1996 WO
WO 9608253 Mar 1996 WO
WO9608253 Mar 1996 WO
WO 9614058 May 1996 WO
W0-9702020 Jan 1997 WO
9703672 Feb 1997 WO
9703673 Feb 1997 WO
9725028 Jul 1997 WO
9732093 Sep 1997 WO
WO9733566 Sep 1997 WO
9814168 Apr 1998 WO
W0-9817261 Apr 1998 WO
W0-9820095 May 1998 WO
9828345 Jul 1998 WO
9833378 Aug 1998 WO
WO 9841194 Sep 1998 WO
9903471 Jan 1999 WO
WO 9920255 Apr 1999 WO
WO 99032119 Jul 1999 WO
WO 9932120 Jul 1999 WO
WO-9932093 Jul 1999 WO
WO99039698 Aug 1999 WO
WO 9944591 Sep 1999 WO
9951209 Oct 1999 WO
W09902142 Jan 2000 WO
W00024383 Apr 2000 WO
0025752 May 2000 WO
0059479 Oct 2000 WO
0059481 Oct 2000 WO
W0-0072847 Dec 2000 WO
WO 0132148 May 2001 WO
0158433 Aug 2001 WO
WO02036099 May 2002 WO
WO 0287512 Jul 2002 WO
WO02092059 Nov 2002 WO
WO04026256 Apr 2004 WO
WO04064807 Aug 2004 WO
WO04084865 Oct 2004 WO
WO04093819 Nov 2004 WO
WO04108117 Dec 2004 WO
W0-2005020929 Mar 2005 WO
WO05034930 Apr 2005 WO
WO 2005041968 May 2005 WO
WO05099674 Oct 2005 WO
W0-20061 09183 Oct 2006 WO
WO07048233 May 2007 WO
WO07103293 Sep 2007 WO
WO07112574 Oct 2007 WO
WO08140460 Nov 2008 WO
2009036287 Mar 2009 WO
WO09036812 Mar 2009 WO
WO09059701 May 2009 WO
WO10033195 Mar 2010 WO
WO 2012131463 Oct 2012 WO
Non-Patent Literature Citations (322)
Entry
Purdue Pharma L.P. v. Recro Technology LLC, Court of Appeals for the Federal Circuit, Case No. 16-2260, Opinion filed and Judgment entered, Document No. 46-1; Jun. 13, 2017.
The Declaration of Benjamin Oshlack filed in U.S. Appl. No. 13/833,263 on Dec. 24, 2014.
Purdue Pharma L.P. v. Recro Technology LLC, Court of Appeals for the Federal Circuit, Case 16-2260, Corrected Brief of Appellant Purdue Pharma, L.P., Oct. 4, 2016.
Purdue Pharma L.P. v. Recro Technology LLC, Court of Appeals for the Federal Circuit, Case 16-2260, Brief for Appellee Recro Technology, LLC, Nov. 14, 2016.
Purdue Pharma L.P. v. Recro Technology LLC, Court of Appeals for the Federal Circuit, Case 16-2260, Reply Brief of Appellant Purdue Pharma, L.P., Dec. 28, 2016.
Physician's Desk Reference, p. 272 (1947).
Physician's Desk Reference, pp. 1144, 1126, 1130, 1144-1145, 2023, 2498, 2499 (1994).
J.W. Barnhart & J. Caldwell, Gas Chromatographic Determination of Hydrocodone in Serum, 130 J. Chromatography, pp. 243, 249 (1977).
Remington: The Science and Practice of Pharmacy, 19th Ed., pp. 1199-1200, 1661 (1995).
Remington: The Science and Practice of Pharmacy, 20th ed., pp. 906, 909, 1446-1448 (2000).
M. Mayersohn and S. Tannenbaum, “On Reclaiming Data from the Literature: Literature Data “R and R” (Recovery and Reanalysis)” with a commentary by Dr. Gerhard Levy, “Using Other People's Data in Publications”, Amer. J. Pharm. Ed., 62, pp. 363-370 (1998).
Principles and Perspectives in Drug Bioavailability, Blanchard et al., p. 62, eds. (1979).
Clinical Pharmacokinetics—Concepts and Applications, M. Rowland and T.N. Tozer eds., p. 100, 3rd ed. (1995).
H. William Kelly and Shirley Murphy, Efficacy of a 12-hour sustained-release preparation in maintaining therapeutic serum theophylline levels in asthmatic children, 66 Pediatrics 97(1980).
P. Macheras et al., Biopharmaceutics of Orally Administered Drugs 30 (1995).
Goodman and Gilman's, The Pharmacological Basis of Therapeutics, pp. 528, 533, 535 (Alfred Goodman Gilman et al., eds. 9th ed. (1996).
Handbook of Pharmaceutical Excipients, pp. 102, 110, 244, 252, 276, 283, 300, 305, 399, 555 (3rd ed. 2000).
Guo et al., Pharmaceutical Applications of Naturally Occurring Water-Soluble Polymers, 1 Pharm. Sci. Tech. Today 254, pp. 254, 257 (1998).
H.G. Kristensen and T. Schaefer, Granulation: A Review on Pharmaceutical Wet Granulation, Drug Development and Industrial Pharmacy 13 (4), pp. 803-872, (1987).
U.S. Pharmacopeia, pp. 1791-93, 1924, 1925, 2053 (23d ed.) (1995).
Oral Drug Products in Oral Drug Absorption—Prediction and Assessment, pp. 200-202 (J.B. Dressman and H. Lennernas eds.), ( 2000).
The Merck Index, pp. 989, 757, 762 (Susan Budavari et al., eds. 11th ed.) (1989).
Goodman and Gilman's, The Pharmacological Basis of Therapeutics, pp. 487, 496, 497 (Alfred Goodman Gilman et al., eds. 8th ed. (1990).
Bailey, Poly(Ethylene Oxide), (1976).
L'Hote-Gaston et al. The Use of Polyethylene Oxide Mixtures to Study Formulation Robustness in Hydrophilic Extended Release Matrix Tablets (2009).
Dow, Degradation of Water-Soluble Resins (2002).
Coppens et al. Thermal and Rheological Evaluation of Pharmaceutical Excipients for Hot Melt Extrusion (2004).
Coppens et al. Hot Melt Extrusion: Effect of Polymer Selection and Processing on Drug Dissolution (2005).
Körner et al., “Molecular Information on the Dissolution of Polydisperse Polymers: Mixtures of Long and Short Poly(ethylene oxide),” J. Phys. Chem. B., 109 (23), 11530-11537 (2005).
Ozeki et al. Mechanism of medicine release from solid dispersion composed of poly(ethylene oxide)-carboxyvinylpolymer interpolymer complex and pH effect on medicine release. Intl. J. Pharm. 171 pp. 123-132 at 124 (1998).
Carraher, Polymer Chemistry (6th ed.) (2003).
Oberlerchner J.T. et al., Overview of Methods for Direct Molar Mass Determination of Cellulose, 20 Molecules 10313-41 (2015).
Manas Chanda and Salil Roy, Plastics Technology Handbook 6 (3d ed.) (1998).
The Office Action issued in connection with Chinese Patent Application No. CN 201210130057.4 dated Oct. 11, 2014.
English Abstract of JP-08157392-A, Jun. 18, 1996.
Hansch, C. et al., “Rational Design, Mechanistic Study and Therapeutic Application of Chemical Compounds,” Medicinal Chemistry, vol. 5, (1990), pp. 251-278.
Ratain M. J. et al., “Population Pharmacodynamic Study of Amonafide: A Cancer and Leukemia Group B Study,” Journal of Clinical Oncology, vol. 13, No. 3, (1995), pp. 741-747.
Abstract of Yasuhara, H., “Ethnic Factors in evaluation of drug efficacy and safety,” Database Medline US National Library of Medicine (NLM) Bethesda, vol. 104 No. 2 (1994), pp. 67-78.
Setiabudy R., et al., “Dapsone N-Acetylation, Metoprolol, Alpha-Hydroxylation Polymorphisms in an Indonesian Population: A cocktail and Extended Phenotyping Assessment Trial,” Clinical Pharmacology & Therapeutics, vol. 56, No. 2, (1994), pp. 142-153.
“Helsinki Declaration,” http://onlineethics.org/reseth/helsinki.html, retrieved on Apr. 5, 2005.
Adv. Drug Deliv. Rev. Mar. 1, 1999; 36(1):125-141.
The Decision to refuse a European Patent application issued in connection with European patent application No. 10 180 984.6-1464 on Jul. 9, 2014.
The Office Action issued in connection with Japanese patent application No. 2013-168584 dated Sep. 16, 2014.
Drugs Jan. (1999); 57(1 ):93-9.
Johnson, Sarah J., “Opioid Safety in Patients With Renal or Hepatic Dysfunction” Pain Treatment Topics pp. 1-9 (Release date Jun. 2007; updated Nov. 30, 2007).
Emami, J. et al, “In vitro-In vivo Correlation: From Theory to Applications”, J. Pharm Pharmaceut Sci, (www.cspsCanada.org) 9 (2), 169-189, 2006.
Lippold, B. C., Constant or Pulsed Delivery of Active Substance?, Pharmacie in uns Zeit, Jan. 13-31, 1990, No. 1.
Clin. Neuropharmacol Jan.-Feb. (1999); 22 (1):33-9).
Clin. Phannacokinet Sep. (1998); 35(3):173-90.
Conte. et al., Drug Del.. Ind. Pharm. (1989) 15:2583-2596.
J. Control Release Aug. 5, 1999; 60(2-3):391-7.
J. Pharm. Pharmacol Dec. (1996); 48(12):1276-84.
J. Pharm. Sci. Feb. (1993); 82(2):113-26.
Zimm et al., Pharmaceutical Development and Technology, 1(1), 37-42 (1996) “Drug Release from a Multiparticulate Pallet System.”
Thomsen, L. Juul, “Prolonged Release Matrix Pellets Prepared by Melt Pelletization. Part IV: Drug Particles Size, and Binder Composition,” Pharmaceutical Technology Europa, pp. 19-24 (Oct. 1994).
The Applicant-Initiated Interview Summary issued in connection with U.S. Appl. No. 13/833,263 dated Nov. 3, 2014.
The Office Action issued in connection with U.S. Appl. No. 13/833,263 dated Oct. 7, 2014.
The Office Action issued in connection with U.S. Appl. No. 13/833,263 dated Jun. 23, 2014.
The Applicant-Initiated Interview Summary issued in connection with U.S. Appl. No. 13/833,263 dated May 12, 2014.
The Office Action issued in connection with U.S. Appl. No. 13/833,263 dated Feb. 6, 2014.
The Advisory Action issued in connection with U.S. Appl. No. 12/982,386 dated Oct. 9, 2014.
The Office Action issued in in connection with U.S. Appl. No. 12/982,386 dated Aug. 13, 2014.
The Office Action issued in in connection with U.S. Appl. No. 12/982,386 dated Feb. 26, 2014.
The Advisory Action issued in connection with U.S. Appl. No. 12/982,386 dated Dec. 19, 2014.
The Office Action issued in in connection with U.S. Appl. No. 12/982,386 dated Sep. 4, 2014.
The Applicant-Initiated Interview Summary issued in connection with U.S. Appl. No. 14/483,395 dated Oct. 3, 2014.
The Office Action issued in connection with U.S. Appl. No. 14/483,395 dated Oct. 9, 2014.
The claims pending in U.S. Appl. No. 14/483,395 on Nov. 17, 2014.
The claims pending in U.S. Appl. No. 13/833,263 on Nov. 17, 2014.
The claims pending in U.S. Appl. No. 12/982,386 on Nov. 17, 2014.
The claims pending in U.S. Appl. No. 14/520,032 on Nov. 17, 2014.
The Decision to refuse a European Patent application issued in connection with European patent application No. 01992565.0-1464 dated Jun. 27, 2014.
Guidance for Industry, Extended Release Oral Dosage Forms: Development, Evaluation, and Application of In Vitro/ In Vivo Correlations; US Dept. of Health and Human Services Food and Drug Administration; Center for Drug Evaluation and Research (CDER), Sep. 1997, pp. 1-24.
Giunchedi et al., J. Pharm. (1991) 77:177-181.
Eur. J. Pharm. Sci. Jul. (1999); 8(3):157-9.
Shah et al., J. Cont. Rel. (1989) 9:169-175.
The Office Action issued in in connection with U.S. Appl. No. 12/982,386 dated Apr. 30, 2013.
The Applicant-Initiated Interview Summary issued in connection with U.S. Appl. No. 12/982,386 dated Apr. 18, 2013.
The Office Action issued in in connection with U.S. Appl. No. 12/982,386 dated Nov. 5, 2012.
The Office Action issued in in connection with U.S. Appl. No. 12/982,386 dated Apr. 10, 2012.
An English translation of the Office Action issued in connection with Israeli Patent Application No. 155,637 dated Nov. 16, 2014.
An English translation of the Decision on Reexamination issued in connection with Chinese patent application No. 200910132824.3 dated Nov. 5, 2014.
An English translation of the Notice on Reexamination issued in connection with Chinese patent application No. 200810125922.X dated Dec. 3, 2014.
Claims pending in U.S. Appl. No. 12/982,386 on Jan. 13, 2015.
Claims pending in U.S. Appl. No. 14/483,3959 on Jan. 13, 2015.
Gourlay, et al., “Once a Day (i.e. 24 Hourly) Kapano™, A New Sustained Release Morphine Formulation, in the Treatment of Cancer Pain: Pharmacokinetic Aspects,” European Journal of Cancer; Part A General Topics 1995:31 (S5) Suppl: S187 Abs 897, European Conference on Clinical Oncology and Cancer Nursing, Paris, Oct. 29-Nov. 2, 1995.
Claims pending in U.S. Appl. No. 13/833,263 on Jan. 5, 2015.
Brendenberg “New Concepts in Administration of Drugs in Tablet Form: Formulation and Evaluation of a Sublingual Tablet for Rapid Absorption and Presentation of an Individualized Dose Administration System Acta Universitiatis Upsaliensis.” Comprehensive Summaries of Uppsa/a Dissertations from the Faculty of Pharmacy 287 83 pp. Uppsala ISBN 91-554;5600-6 (2003).
Frohof-Hulsmann et al., “Aqueous Ethyl Cellulose Dispersion Containing Plasticizers of Different Water Solubility and Hydroxypropyl Methyl-Cellulose as Coating Material for Diffusion Pellets II: Properties of Sprayed Films”, European Journ. of Pharma and Biopharma., vol. 48, pp. 67-75, 1999.
Gustafsson et al., “Characterisation of Particle Properties and Compaction Behaviour of Hydroxypropyl Methylcellulose with Different Degrees of Methoxy/Hydroxypropyl Substitution”, EP Journ of Pharmaceutical Sci. 9, pp. 171-184, 1999.
Hyppola et al.,“Evaluation of Physical Properties of Plasticized Ethyl Cellulose Films Cast From Ethanol Solution Part 1”, International Journ. of Pharma., vol. 133, pp. 161-170, 1996.
Physician's Desk Reference 57th ed. 2003 p. 1184-1185 (package insert information for ACTIQ).
Portenoy et al. “Fentanyl Buccal Tablet (FBT) for Relief of Breakthrough Pain in Opioid-Treated Patients with Chronic Low Back Pain: A Randomized, Placebo-Controlled Study”, ASRA 06, Final Abstract, Submitted Aug. 4. 2007.
Portenoy et al. “Fentanyl Buccal Tablet (FBT) for Relief of Breakthrough Pain in Opioid-Treated Patients with Chronic Low Back Pain”, Current Medical Research and Opinion, vol. 23(7), pp. 223-233, 2007.
Siepmann et al., “A New Model Describing the Swelling and Drug Release Kinetics from Hydroxypropyl Methylcellulose Tablets”, Journ. of Pharmaceutical Sciences, vol. 88, No. 1, pp. 65-72, Jan. 1999.
Sung et al., “Effect of Formulation Variables on Drug and Polymer Release from HPMC-Based Matrix Tablets”, International Journ. of Pharmaceutics vol. 142, pp. 53-60, 1996.
Vashi et al., “Clinical Pharmacology and Pharmacokinetics of Once-Daily Hydromorphone Hydrochloride Extended-Release Capsules”, J. Clin. Pharmacal, vol. 45, pp. 547-554, 2005.
Viriden et al., “Investigation of Critical Polymer Properties for Polymer Release and Swelling of HPMC Matrix Tablets”, EP Journal of Pharmaceutical Sciences 36, pp. 297-309, 2009.
Webster, PTI-821: Sustained-Release Oxycodon Investig. Drugs, vol. 16, (3), pp. 1-8, 2007.
Claims pending in U.S. Appl. No. 14/581,175 on Jan. 5, 2015.
International Preliminary Report on Patentability and Written Opinion re: PCT/US2011/035767 dated Nov. 13, 2012.
International Preliminary Report on Patentability and Written Opinion re: PCT/US2011/035768 dated Nov. 13, 2012.
International Preliminary Report on Patentability and Written Opinion re: PCT/US2011/035770 dated Nov. 13, 2012.
International Preliminary Report on Patentability and Written Opinion re: PCT/US2011/025914 dated Aug. 28, 2012.
International Search Report and Written Opinion for PCT/US2007/020041, dated Feb. 25, 2008.
“The Merck Manual,” Merck and Co., p. 4711 (1989).
English Translation of the Office Action dated Mar. 22, 2011 in connection with Japanese Patent Application No. 2006-3165067.
European Search Report issued in connection with European Application No. 10177508.8-2123 dated Sep. 11, 2010.
Encarta World English Dictionary [North American Edition] 2006 Microsoft Incorporation “Matrix”, p. 1, definition 2, http://encarta.msn.com/encnet/features/dictionary/DictionaryResults.aspx?- refid=1861678516 retrieved on Apr. 30, 2006.
Wiktionary, “dispersed”, p. 1, http://en.wiktionary.org/wiki/disperse retrieved on Apr. 30, 2006.
Castensen et al., “USP Dissolution IV: Comparison of Methods” J. Pham. Sci., 67(9), pp. 1303-1307, (1978).
Santus G., and Baker. R.W., “Osmotic drug delivery: a review of the patent literature”, J Controlled Release (1995) vol. 35 pp. 1-21.
Hansch et al., “Comprehensive Medicinal Chemistry, vol. 5, Biopharmaceutics,” Oxford, Pergamon Press, 1990, pp. 251-278.
Ratain et al., “Population pharmacodynamic study of amonafide: A cancer and leukemia group B study,” Journal of Clinical Oncology, Grune and Stratton, 1995, pp. 741-747.
Database Medline Online! US National Library of Medicine, Yasuhara “Ethnic factors in evaluation of drug efficacy and safety”, 1994.
Setiabudi et al., “Dapsone N-acetylation, Metoprolol, alpha-hydroxylation, and 5-mephenytoin 4-hydroxylation polymorphisms in an Indonisian population: A cocktail and extended phenotyping assessment trial,” Clinical Pharmacology & Therapeutics, 1994, pp. 141-153.
Database internet “Online!” Helsinki Declaration, 2002.
English translation of Office Action issued by Japanese Patent Office in connection with corresponding Japanese Patent Application No. 2001-534353 dated May 25, 2010.
Merk Index, 13th edition, entry 4806: hydrocodone (2001).
EPI Journal Feb. 2007, pp. 59-60; David Harrison; “Divisional Application a continuing problem” (2007).
Document CA/PL 17/07; “Misuse of Divisional Applications,” by the EPO President, addressed to the EPO Committee of Patent Law. Presented at the EPO Administrative Council, dated Oct. 5, 2007.
Document CA/PL 3/08, presented by epi and other Professional Representatives, addressed to the EPO Committee of Patent Law. Presented at the EPO Administrative Council, dated Feb. 14, 2008.
Summons to Attend Oral Proceedings issued in connection with European Patent Application No. 05019453.9 on Jul. 23, 2010.
Abraham Sunshine, et al., “Analgesic oral efficacy of tramadol hydrochloride in postoperative pain,” Clin. Pharmacol. Ther., Jun. 1992, pp. 740-746.
E. Beubler, “Medikamentose Schmerztherapie: Kriterien, Moglichkeiten, Risken,” Therapiewoche Osterreich, 7, 2 (1992), pp. 1-15, English translation.
Geoffrey K. Gourlay, Ph.D., et al., “Influence of a high-fat meal on the absorption of morphine from oral solutions,” Clin. Pharmacol. Ther., Oct. 1989, pp. 463-468.
Robert Kaiko, et al., “A Single-Dose Study of the Effect of Food Ingestion and Timing of Dose Administration on the Pharmacokinetic Profile of 30-MG Sustained-Release Morphine Sulfate Tablets,” Current Therapeutic Research, vol. 47, No. 5, May 1990, pp. 869-878.
Geoffrey K. Gourlay, Ph.D., “The Reproducibility of Bioavailability of Oral Morphine from Solution Under Fed and Fasted Conditions,” Journal of Pain and Sympton Management, vol. 6., No. 7, Oct. 1991, pp. 431-436.
Robert F. Kaiko, et al., “Controlled-Release Morphine Bioavailability (MS Contin Tablets) in the Presence and Absence of Food,” The Hospice Journal, vol. 6(4) 1990, pp. 17-30.
N. Yokokawa, et al., “Relationship between plasma concentration of morphine and analgesic effectiveness,” Postgrad Med J, (1991) 67 (Suppl. 2) pp. S50-S54.
Physicians Desk Reference 1994, 48th Edition, pp. 1821-1824.
D.L. Munday, “Changes in Drug Release Rate 2 Effect of Temperature and Relative Humidity on Polymeric Film Coatings,” 5th Cong. Int. Tech. Pharm., 1989, vol. 2, pp. 55-60.
A Protocol for a clinical study entitled “A Randomized, Double-Blind, Parallel-Group Study comparing the Efficacy and Safety of Kapanol® to MsContin® in the Management of Patients with Moderate to Severe Cancer Pain” (“The Protocol”). The date of the Protocol is indicated as Feb. 10, 1992 and it bears COD No. 14556. The sponsor of the study is indicated to be Faulding Pharmaceuticals, an Australian company.
Certain Patient Diary Cards, Drug Disposition Records, Case Reports Forms and a listing which apparently correlates patient randomization number with the treatment of dosing regimen assigned to each patient, 1992.
Patient consent forms, apparently for four study participants. cited by other . Investigator Agreements between the study organizers and certain of the principal investigators, 1992.
Abstracts from the Twelfth Annual Congress of the Oncology Nursing Society, May 1987.
Lapin et al., “Cancer Pain Management with a Controlled Release Oral Morphine Preparation,” Journ. of Pain and Sympton Manag., v4 (3), pp. 146-151, 1989.
J. Lapin et al., “Guidelines for use of Controlled Release Oral Morphine in Cancer pain Management,” Cancer Nursing, v 12 (4), pp. 202-208, 1989.
R.F. Kaiko, “The Pre-and Postoperative Use of Controlled-Release Morphine (MS Contin Tablets): A Review of the Published Literature,” Medical Department, The Purdue Frederick Company, Royal Society of Medical Services, International Congress, Symposium Services, No. 149, pp. 147-160 (1989).
H.F. Slowey et al., “Effect of Premedication with Controlled-Release Oral Morphine on Postoperative Pain,” Anaesthesia, 1985, vol. 40, pp. 438-440. cited by other .MS Contin—Frequency of Daily Dosing, Jan.-Nov. 1990.
R.K. Portenoy, et al., “A Randomized, Double-Blind, Double-Dummy, Crossover Study Comparing the Pharmacokinetics and Pharmacodynamics of Kapanol® Capsules Given Every 24 hours and Every 12 hours with MS Contin® Tablets Given Every 12 Hours in the Management of Patients with Moderate to Severe Chronic Pain” (1992).
R.West et al., World Congress on Pain Abstracts 997-1001, Aug. 26, 1993.
Advertisement: Roxanol SR., 1988 Roxane Labs, Inc.
R. Kaiko and T. Hunt, Clip. Thera. vol. 13, No. 4, pp. 484-488, 1991.
S. Bloomfield, et al. Clin. Pharmacol. Ther. vol. 53, No. 4, pp. 469-478, 1993.
Advertisement: MS Contin 1986, 1987 the Purdue Frederick Company.
Sustained Release Medications, Noyes Data Corp., pp. 3,4, 10-15, 96-99, 335-337 (1980).
Flanders, P., et al., “The Control of Drug Release From Conventional Melt Granulation Matrices,” Drug Development and Industrial Pharmacy, vol. 13, No. 6, pp. 1001-1022 (1987).
McTaggart, Celia M., et al., “The evaluation of formulation and processing conditions of a melt granulation process,” International Journal of Pharmaceutics, vol. 19, pp. 139-148 (1984).
Schaefer, T., et al., “Melt granulation in a laboratory scale high shear mixer,” Drug Development and Industrial Pharmacy, vol. 16, No. 8., pp. 1249-1277 (1990).
Thomsen, L. Juul, et al., “Prolonged Release Matrix Pellets Prepared by Melt Pelletization I. Process Variables,” Drug Development and Industrial Pharmacy, vol. 19, No. 15, pp. 1867-1887 (1993).
Thomsen, L. Juul, et al., “Prolonged Release Matrix Pellets prepared by Melt Pelletization II. Hydrophobic Substances as Meltable Binders,” Drug Development and Industrial Pharmacy, vol. 20, No. 77, pp. 1179-1197 (1994).
Thomsen, L. Juul, “Utilizing melt pelletization technique for the preparation of prolonged release products,” Pelletization, (material elaborated by assistant prof. Lars Juul Thomsen, Department of Pharmaceutics, Royal Danish School of Pharmacy for the DIE course “Pelletization Technologh,” Nov. 1992, 106 pages plus 3 appendices.
Maccarrone C. et al.; Single Dose Pharmacokinetics of Kapanol.TM. , a New Oral Sustained-Release Morphine Formulation; Clinical Drug Investigation 1994:7 (5) 262-274.
West R. J. Maccarrone C. Single dose pharmacokinetics of a new oral sustained release morphine formulation, Kapanol® capsules. (Abstract 997) International Association for the Study of Pain, 7th World Congress on Pain. Paris, Aug. 22-27, 1993 (Data on file, Glaxo Australia, F.H. Faulding).
Gourlay GK, Plummer JL, Cherry DA, et al. A comparison of Kapanol.TM. (A new sustained release morphine formulation), MST Continus® and morphine solution in cancer patients: pharmacokinetic aspects. (Abstract 998) International Association for the Study of Pain, 7.sup.th World Congress on Pain. Paris, Aug. 22-27, 1993 (Data on file, Glaxo Australia, F.H. Faulding).
Cherry DA, Gourley GK, Plummer JL, et al. A comparison of Kapanol™ (a new sustained release morphine formulation), MST Continus® and morphine solution in cancer patients: morphine metabolite profiles and renal function. (Abstract 999) International Association for the Study of Pain, 7th World Congress on Pain, Paris, Aug. 22-27, 1993 (Data on file, Glaxo Australia, F.H. Faulding).
Plummer JL, Cherry DA, Gourlay GK, et al. A comparison of Kapanol™ (a new sustained release morphine formulation) MST Continus® and morphine solution in cancer patients: pharmacodynamic aspects. (Abstract 1000) International Association for the Study of Pain, 7th World Congress on Pain, Paris, Aug. 22-27, 1993 (Data on file, Glaxo Australia, F.H. Faulding).
Toner G, Cramond T, Bishop, et al. Randomized double blind, phase III crossover study of a new sustained-release oral morphine formulation, KapanolTM capsules, (Abstract 1001) International Association for the Study of Pain, 7th World Congress on Pain, Paris, Aug. 22-27, 1993 (Data on file, Glaxo Australia, F.H. Faulding).
European Journal of Cancer; Once a Day (i.e. 24 Hourly) KapanolTM, A New Sustained Release Morphine Formulation, in the Treatment of Cancer Pain: Morphine Metabolite Profiles; Part A General Topics 1995; 31 (S5) Suppl:S184 Abs 884, European Conference on Clinical Oncology and Cancer Nursing, Paris, Oct. 29-Nov. 2, 1995.
European Journal of Cancer; Kadian™/Kapanol™—A Once Daily Morphine Formulation; Part A General Topics 1995; 31 (S5) Suppl: S182 Abs 873 European Conference on Clinical Oncology and Cancer Nursing, Paris, Oct. 29-Nov. 2, 1995.
Gourlay et al., Proceeding of the 7th World Congress on Pain; A comparison of Kapanol (a New Sustained-Release Morphine Formulation), MST Continus, and Morphine Solution in Cancer Patients: Pharmacokinetic Aspects of Morphine and Morphine Metabolites Progress in Pain Research and Management vol. 2 pp. 631-643, 1993.
Kaiko R.F.; Clinical Protocol and Role of Controlled Release Morphine in the Surgical Patient; Anesthesiology and Pain Management 1991, pp. 193-212.
MS Contin—Frequency of Daily Dosing (NDTI)—Jun. 1991-May 1992.
The Merck Index, eleventh edition, pp. 757 and 1100 (1989).
MS Contin—Frequency of Daily Dosing, Jan.-Nov.1990.
Encyclopedia of Controlled Drug Delivery, vol. 1, “In vitro-in vivo correlation,” pp. 425- 435 (1999).
Encyclopedia of Controlled Drug Delivery, vol. 2, “Oral drug delivery, small intestine & colon,” pp. 698-728 (1999).
Guidance for Industry, “Extended Release Oral Dosage Forms: Development, Evaluation, and Application of In Vitro/ In vivo Correlation” ( FDA, 1997).
Opinion Expressed by the Board of Appeal in the Summons to Oral Proceedings in Case EP02026247 ( May 24, 2011).
Goodman and Gilman's, The Pharmacological Basis of Therapeutics, pp. 7-8 (1990).
Office Action dated May 24, 2011, in connection with European Application No. 10 179 087.1-2123.
Office Action dated Jul. 27, 2012, in connection with European Application No. 01 992 565.0-2123.
Office Action dated Jul. 27, 2012, in connection with European Application No. 10 177 508.8-2123.
Office Action dated Jul. 27, 2012, in connection with European Application No. 10 179 086.3-2123.
Office Action dated Jul. 27, 2012, in connection with European Application No. 10 180 945.7.
Office Action dated Jul. 27, 2012, in connection with European Application No. 10 180 984.6.
Office Action dated Jul. 27, 2012, in connection with European Application No. 10 181 032.3.
Zohydro ER Package Insert revised Oct. 2013.
Advisory Committee Briefing Document, NDA 20-2880, Zohydro™ ER Hydrocodone Bitartrate Extended-Release Capsules, Anesthetic and Analgesic Drug Products Advisory Committee, Dec. 7, 2012.
FDA Response to a Citizen Petition, Oct. 25, 2013.
Summons to attend oral proceedings issued on Jun. 5, 2013, in connection with European Patent Application No. 10180984.6.
Summons to attend oral proceedings issued on Jun. 5, 2013, in connection with European Patent Application No. 10181032.3.
Summons to attend oral proceedings issued on Jun. 5, 2013, in connection with European Patent Application No. 10177508.8.
Summons to attend oral proceedings issued on Jun. 5, 2013, in connection with European Patent Application No. 01992565.0.
Summons to attend oral proceedings issued on Jun. 5, 2013, in connection with European Patent Application No. 10179087.1.
Summons to attend oral proceedings issued on Jun. 5, 2013, in connection with European Patent Application No. 10179086.3.
Claims pending in U.S. Appl. No. 13/901,069 on Nov. 22, 2013.
Claims pending in U.S. Appl. No. 13/901,761 on Nov. 22, 2013.
Claims pending in U.S. Appl. No. 12/982,386 on Nov. 22, 2013.
Approval Package for NDA 20-616/5-001 (Kadian®), 1997.
Kadian® Prescribing Information 2006.
MS Contin® Prescribing Information 2012.
Oxycontin® Prescribing Information 2010.
Trandemate ER® PrescribingInformation 2005.
The Office Action issued in connection with U.S. Appl. No. 12/982,386 dated Feb. 26, 2014.
Benziger, et al., “A Pharmacokinetic/Pharmacodynamic Study of Controlled-Release Oxycodone”, 1997.
Claims pending in U.S. Appl. No. 13/833,263 on Dec. 9, 2013.
Zohydro ER Hydrocodone Bitartrate Extended-Release (HC-ER) Dec. 7, 2012.
The Office Action issued in connection with U.S. Appl. No. 13/833,263, dated Jun. 23, 2014.
The Office Action issued in connection with U.S. Appl. No. 14/094,968, dated Jun. 23, 2014.
The Office Action issued in connection with U.S. Appl. No. 12/982,386, dated Aug. 13, 2014.
Santus G., et al., “Osmotic drug delivery: a review of the patent literature,” Journal of Controlled Release, vol. 35, No. 1, (1995), pp. 1-21.
M.H. Beers: “The Merck Index, 11th edition,” 1989, Merck & Co. CP002606249.
European Search Report dated Nov. 9, 2010 in connection with European Application No. 10180945.7-2123.
Claims pending in U.S. Appl. No. 14/210,565 on Mar. 10, 2015.
Claims pending in U.S. Appl. No. 14/612,483 on Mar. 10, 2015.
Amendment after final action under 37 C.F.R. § 1.116 filed in connection with U.S. Appl. No. 11/372,857 dated Mar. 2, 2015.
Declaration under 37 C.F.R. § 1.132 by Gurvinder Singh Rekhi filed in connection with U.S. Appl. No. 11/372,857 on Mar. 6, 2015.
Declaration of Interference issued in connection with U.S. Appl. Nos. 11/372,857; 13/833,263 and 14/094,968 on Apr. 8, 2015.
Claims pending in U.S. Appl. No. 14/635,198 on Apr. 14, 2015.
Claims pending in U.S. Appl. No. 14/672,894 on Apr. 14, 2015.
Waiver of In Vivo Bioavailability and Bioequivalence Studies for Immediate Release Solid Oral Dosage Forms Containing Certain Active Moieties/Active Ingredients Based on a Biopharmaceutics Classification System (Issued Jan. 1999, Posted Feb. 16, 1999).
J. W. Barnhart, .et. al., Gas Chromatographic Determination of Hydrocodone in Serum, 130 J. Chromatography 243-49, Abstract (1977).
Interference 106,022, Paper 6 Oshlack Notice of Related Proceedings, Apr. 22, 2015.
Interference 106,022, Paper 12 John G. Devane Notice of Related Proceedings, Apr. 22, 2015.
Interference 106,022, Paper 14 Redeclaration 37 C.F.R. 203(c), Apr. 23, 2015.
Interference 106,022, Paper 18 Purdue Pharma Motion List, May 7, 2015.
Interference 106,022, Paper 19 Recro Motion List, May 8, 2015.
Interference 106,022, Paper 20 Recro Statement Concerning Other Proceedings, May 8, 2015.
Interference 106,022, Paper 21 Order Authorizing Motions and Setting Times, May 19, 2015.
Interference 106,022, Paper 22 Redeclaration 37 C.F.R. 41.203(c), May 19, 2015.
Interference 106,022, Paper 25 Decision Miscellaneous Motion Bd R 121(a)(3), Jul. 2, 2015.
Interference 106,022, Paper 148 Recro Motion 3, Jul. 10, 2015.
Interference 106,022, Paper 123 Recro Motion 1, Jul. 10, 2015.
Interference 106,022, Paper 140 Recro Motion 5, Jul. 10, 2015.
Interference 106,022, Paper 27 Recro Exhibit List, Jul. 10, 2015.
Interference 106,022, Paper 142 Purdue Pharma Exhibit List, Jul. 10, 2015.
Interference 106,022, Paper 143 Purdue Pharma Motion 1, Jul. 10, 2015.
Interference 106,022, Paper 144 Purdue Pharma Motion 2, Jul. 10, 2015.
Interference 106,022, Paper 145 Purdue Pharma Motion 4, JMuly 10, 2015.
Interference 106,022, Paper 152 Order—Responsive Motion, Jul. 24, 2015.
Interference 106,022, Paper 154 Purdue Motion 5, Jul. 31, 2015.
Interference 106,022, Paper 163 Recro Motion 6, Jul. 31, 2015.
Interference 106,022, Paper 159 Recro Updated Exhibit List, Jul. 31, 2015.
Interference 106,022, Exhibit 2001, Jul. 10, 2015.
Interference 106,022, Exhibit 2002, Jul. 10, 2015.
Interference 106,022, Exhibit 2003, Jul. 10, 2015.
Interference 106,022, Exhibit 2004, Jul. 10, 2015.
Interference 106,022, Exhibit 2005, Jul. 10, 2015.
Interference 106,022, Exhibit 2006, Jul. 10, 2015.
Interference 106,022, Exhibit 2007, Jul. 10, 2015.
Interference 106,022, Exhibit 2008, Jul. 10, 2015.
Interference 106,022, Exhibit 2009, Jul. 10, 2015.
Interference 106,022, Exhibit 2010, Jul. 10, 2015.
Interference 106,022, Exhibit 2011, Jul. 10, 2015.
Interference 106,022, Exhibit 2012, Jul. 10, 2015.
Interference 106,022, Exhibit 2013, Jul. 10, 2015.
Interference 106,022, Exhibit 2014, Jul. 10, 2015.
Interference 106,022, Exhibit 2015, Jul. 10, 2015.
Interference 106,022, Exhibit 2016, Jul. 10, 2015.
Interference 106,022, Exhibit 2017, Jul. 10, 2015.
Interference 106,022, Exhibit 2018, Jul. 10, 2015.
Interference 106,022, Exhibit 2021, Jul. 10, 2015.
Interference 106,022, Exhibit 2022, Jul. 10, 2015.
Interference 106,022, Exhibit 2023, Jul. 10, 2015.
Interference 106,022, Exhibit 2025, Jul. 10, 2015.
Interference 106,022, Exhibit 2026, Jul. 10, 2015.
Interference 106,022, Exhibit 2027, Jul. 10, 2015.
Interference 106,022, Exhibit 2028, Jul. 10, 2015.
Interference 106,022, Exhibit 2029, Jul. 10, 2015.
Interference 106,022, Exhibit 2030, Jul. 10, 2015.
Interference 106,022, Exhibit 2041, Jul. 10, 2015.
Interference 106,022, Exhibit 2064, Jul. 10, 2015.
Interference 106,022, Exhibit 2065, Jul. 31, 2015.
Interference 106,022, Exhibit 2066, Jul. 31, 2015.
Interference 106,022, Exhibit 2067, Jul. 31, 2015.
Interference 106,022, Exhibit 2068, Jul. 31, 2015.
Interference 106,022, Exhibit 2069, Jul. 31, 2015.
Interference 106,022, Exhibit 2070, Jul. 31, 2015.
Interference 106,022, Exhibit 2071, Jul. 31, 2015.
Interference 106,022, Exhibit 1001, Jul. 10, 2015.
Interference 106,022, Exhibit 1002, Jul. 10, 2015.
Interference 106,022, Exhibit 1003, Jul. 10, 2015.
Interference 106,022, Exhibit 1004, Jul. 10, 2015.
Interference 106,022, Exhibit 1005, Jul. 10, 2015.
Interference 106,022, Exhibit 1006, Jul. 10, 2015.
Interference 106,022, Exhibit 1007, Jul. 10, 2015.
Interference 106,022, Exhibit 1008, Jul. 10, 2015.
Interference 106,022, Exhibit 1009, Jul. 10, 2015.
Interference 106,022, Exhibit 1010, Jul. 10, 2015.
Interference 106,022, Exhibit 1011, Jul. 10, 2015.
Interference 106,022, Exhibit 1012, Jul. 10, 2015.
Interference 106,022, Exhibit 1013, Jul. 10, 2015.
Interference 106,022, Exhibit 1014, Jul. 10, 2015.
Interference 106,022, Exhibit 1015, Jul. 10, 2015.
Interference 106,022, Exhibit 1016, Jul. 10, 2015.
Interference 106,022, Exhibit 1017, Jul. 10, 2015.
Interference 106,022, Exhibit 1019, Jul. 10, 2015.
Interference 106,022, Exhibit 1020, Jul. 10, 2015.
Interference 106,022, Exhibit 1021, Jul. 10, 2015.
Interference 106,022, Exhibit 1022, Jul. 10, 2015.
Interference 106,022, Exhibit 1023, Jul. 10, 2015.
Interference 106,022, Exhibit 1024, Jul. 10, 2015.
Interference 106,022, Exhibit 1025, Jul. 10, 2015.
Interference 106,022, Exhibit 1026, Jul. 10, 2015.
Interference 106,022, Exhibit 1027, Jul. 10, 2015.
Interference 106,022, Exhibit 1028, Jul. 10, 2015.
Interference 106,022, Exhibit 1029, Jul. 10, 2015.
Interference 106,022, Exhibit 1030, Jul. 10, 2015.
Interference 106,022, Exhibit 1041, Jul. 10, 2015.
Interference 106,022, Exhibit 1042, Jul. 10, 2015.
Interference 106,022, Exhibit 1043, Jul. 10, 2015.
Interference 106,022, Exhibit 1044, Jul. 10, 2015.
English translation of the Office Action issued in connection with Japanese Application No. 2014-182108 dated Aug. 11, 2015.
Claims pending in U.S. Appl. No. 14/706,699 on Aug. 21, 2015.
Claims pending in U.S. Appl. No. 14/727,985 on August21, 2015.
Claims pending in U.S. Appl. No. 14/727,997 on August21, 2015.
Claims pending in U.S. Appl. No. 14/728,023 on August21, 2015.
Interference 106,022, Paper 243: Decision on Motions- 37 C.F.R. § 41.127, Apr. 29, 2016.
Mary Eagleson, Polyethylene Glycol, in Concise Encyclopedia Chemistry, p. 865 (1994).
Handbook of Pharmaceutical Excipients, pp. 253-255, 3rd ed. (2000).
Handbook of Pharmaceutical Excipients, pp. 393-400, 3rd ed.( 2000).
Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products—General Considerations (2002).
Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products—General Considerations, p. 13 (2002).
FDA, Guidance for Industry Q1A(R2) Stablity Testing of New Drug Substances and Products (2003).
FDA, Guidance for Industry Q1A(R2) Stablity Testing of New Drug Substances and Products, pp. 1-2 (2003).
The Merck Index, eleventh edition, p. 762 (1989).
Handbook of Pharmaceutical Excipients, pp. 138-140, 209-213 (1986).
Handbook of Pharmaceutical Excipients, pp. 522-524 (2009).
Gilman et al., The Pharmacological Basis of Therapy, 8th ed. p. 497 (1990).
Remington's Pharmaceutical Sciences, 17th ed. (1985) at 1104.
Related Publications (1)
Number Date Country
20170136006 A1 May 2017 US
Provisional Applications (1)
Number Date Country
60162541 Oct 1999 US
Continuations (8)
Number Date Country
Parent 15134901 Apr 2016 US
Child 15376868 US
Parent 14672894 Mar 2015 US
Child 15134901 US
Parent 14635198 Mar 2015 US
Child 14672894 US
Parent 14094968 Dec 2013 US
Child 14635198 US
Parent 13833263 Mar 2013 US
Child 14094968 US
Parent 12982386 Dec 2010 US
Child 13833263 US
Parent 10864829 Jun 2004 US
Child 12982386 US
Parent 09702283 Oct 2000 US
Child 10864829 US