throbber
OXYMORPHONE CONTROLLED RELEASE FORMULATIONS
`
`EL618698014US
`
`BACKGROUND OF THE INVENTION
`
`Pain is the most frequently reported symptom and it is a common clinical problem which
`
`confronts the clinician. Almost 100 million people in the USA suffer from severe pain that,
`
`according to numerous recent reports, is chronically undertreated or inappropriately managed.
`
`The clinical usefulness of the analgesic properties of opioids has been recognized for centuries,
`
`and morphine and its derivatives have been widely employed for analgesia for decades in a
`
`variety of clinical pain states.
`
`Oxymorphone HC1 (14-hydroxydihydromorphinone hydrochloride) is a semi-synthetic
`
`phenanthrene-derivative opioid agonist, widely used in the treatment of acute and chronic pain,
`
`with analgesic efficacy comparable to other opioid analgesics. Oxymorphone is currently
`
`marketed as an injection (1 mg/mL in 1 mL ampules; 1 5 mg/mL in 1 mL ampules; 1.5 mg/mL in
`
`10 mL multiple dose vials) for intramuscular, subcutaneous, and intravenous administration, and
`
`as 5 mg rectal suppositories. At one time, a 10 mg oral immediate release (IR) tablet formation
`
`of oxymorphone HC1 was marketed. Oxymorphone HC1 is metabolized principally in the liver
`
`and undergoes conjugation with glucuronic acid and reduction to 6 alpha and beta hydroxy
`
`epimers.
`
`An important goal of analgesic therapy is to achieve continuous relief of chronic pain.
`
`Regular administration of an analgesic is generally required to ensure that the next dose is given
`
`before the effects of the previous dose have worn off Compliance with opioids increases as the
`
`required dosing frequency decreases. Non-compliance results in suboptimal pain control and
`
`poor quality of life outcomes. (Ferrell B et al. Effects of controlled-release morphine on quality
`
`of life for cancer pain. Oncol Nur Forum 1989;4:521-26). Scheduled, rather than as needed
`
`administration of opioids is currently recommended in guidelines for their use in chronic non-
`
`malignant pain. Unfortunately, evidence from prior clinical trials and clinical experience
`
`suggests that the short duration of action of IR oxymorphone would necessitate 4-hourly
`
`administration in order to maintain optimal levels of analgesia in chronic pain.
`
`935090_1
`
`ENDO - Ex. 2085
`Amneal v. Endo
`IPR2014-00360
`
`

`

`Oshlack et al US Patent 5,266,331 discloses oxycodone controlled release (CR)
`
`formulations. The oxycodone is incorporated into a CR matrix comprising (a) hydrophilic
`
`polymers, such as gum, cellulous ether, acrylic resins and protein-derived materials, (b)
`
`digestible hydrocarbons such as fatty acids, fatty alcohols, etc., and (c) polyalkylene glycols.
`
`The formulation can contain a hydrophobic polymer such as ethyl cellulose. Oxycodone can
`
`also be incorporated into an IR formulation then coated with a CR film. The Oshlack patent
`
`states that other analgesics may be used in place of oxycodone, including hydromorphone,
`
`dihydrocodeine, codeine, dihydromorphine, morphine, buprenorphine, and the like.
`
`ChasM et al. US 5,958,459 discloses CR oral solid formulations of opioid analgesics.
`
`The Chasin patent states that the preferred opioids include mu-agonist opioid analgesics such as
`
`hydromorphone, oxycodone, morphine, levorphanol, methadone, meperidine, heroin,
`
`dihydrocodeine, codeine, dihydromorphine, buprenorphine, and the like. The formulation can be
`
`used in the form of granules, spheroids or pellets in a capsule or in any other suitable solid form.
`
`Other forms mentioned are tablets, microspheres, seeds, ion exchange resin beads, and other
`
`multiparticulate systems. Spheroids or other substrate containing the opioid (e.g. tablet core or
`
`inert pharmaceutical beads) are coated with a CR film of a hydrophobic cellulosic or acrylic
`
`polymer. Beads coated with opioid are prepared by dissolving the opioid in water then spraying
`
`the solution onto the substrate to coat it. The beads are then coated with hydrophobic polymer
`
`and curd to obtain a stabilized release rate.
`
`Sackler et al. US Patent 5,478,577 discloses 24 hour oral opioid formulations that provide
`
`a rapid rate of initial rise of plasma drug level. Oxymorphone is included in a long list of opioids
`
`which can be used. Where the extended release of opioid is due to a CR coating, an IR
`
`formulation can be overcoated on top of the CR coating. When the extended release is due to a
`
`CR matrix, the IR layer can be coated onto the surface of the substrate. Alternatively, a capsule
`
`can be provided that contains both CR particles, e.g., pellets, spheres, beads and the like, and IR
`
`powder or granulate, or a capsule containing CR particles can be coated with an IR formulation.
`
`Suitable materials mentioned in the Sackler patent for inclusion in a CR matrix are (a)
`
`hydrophilic polymers, such as gums, cellulose ethers and acrylic resins, (b) hydrocarbons such as
`
`fatty acids, fatty alcohols, mineral and vegetable oils and waxes, and (c) polyalkylene glycols.
`
`2
`
`935090_1
`
`

`

`Oshlack et al. US Patent 5,958,452 discloses opioid CR formulations made by melt
`
`extrusion of a mixture of opioid, a hydrophobic material, and a retardant material. The Oshlack
`
`patent states that in certain preferred embodiments, the opioid is selected from a list that includes
`
`oxymorphone as well as morphine, codeine, hydromorphone, hydrocodone, oxycodone,
`
`dihydrocodeine, dihydromorphone and tramadol.
`
`A CR formulation of morphone has been demonstrated to provide patients fewer
`
`interruptions in sleep, reduced dependence on caregivers, improved compliance, enhanced
`
`quality of life outcomes, and increased control over the management of pain. In addition, the CR
`
`formulation of morphine provided more constant plasma concentration and clinical effects, less
`
`frequent peak to trough fluctuations, reduced dosing frequency, and possibly fewer side effects.
`
`(Thirlwell MP et al. Pharmacokinetics and clinical efficacy of oral morphine solution and
`
`controlled-release morphine tablets in cancer patients. Cancer 1989; 63:2275-83; Goughnour BR
`
`et al. Analgesic response to single and multiple doses of controlled-release morphine tablets and
`
`morphine oral solution in cancer patients. Cancer 1989; 63:2294-97; Ferrell B et al. Effects of
`
`controlled-release morphine on quality of life for cancer pain. Oncol Nur Forum 1989; 4:521-26.
`
`Oxymorphone IR exhibits low oral bioavailability, because oxymorphone is extensively
`
`metabolized in the liver. Prior to this invention, it was expected that an oxymorphone CR oral
`
`dosage form would also exhibit very low oral bioavailability, and thus would not be useful for
`
`treatment of pain, because it was known that metabolism of drugs that are extensively
`
`metabolized in the liver increases with decreasing release rate.
`
`SUMMARY OF THE INVENTION
`
`To overcome the difficulties associated with a 4-6 hourly dosing frequency of
`
`oxymorphone, this invention provides an oxymorphone CR oral solid dosage form comprising an
`
`analgesically effective amount of oxymorphone or a pharmaceutically acceptable salt of
`
`oxymorphone. Surprisingly, it has been found that the decreased rate of release of oxymorphone
`
`from the oral CR formulation of this invention does not substantially decrease the bioavailability
`
`of the drug as compared to a solution of oxymorphone administered orally. The bioavailability is
`
`sufficiently high that the CR dosage can be used to treat patients suffering moderate to severe
`
`3
`
`935090_1
`
`

`

`pain with once or twice daily dosing. The dosing form of the present invention can also be used
`
`with thrice daily dosing.
`
`The CR dosage form of this invention exhibits a dissolution rate in vitro, when measured
`
`by USP Procedure Drug Release USP 24, of about 25% to about 55% by weight oxymorphone
`
`released after 1 hour, about 55% to about 85% by weight oxymorphone released after 4 hours,
`
`and at least about 80% by weight oxymorphone released after 8 hours .
`
`When administered orally to humans the CR dosage form of this invention exhibits the
`
`following in vivo characteristics: (a) peak plasma level of oxymorphone occurs within about 2 to
`
`about 6 hours after administration; (b) duration of oxymorphone analgesic effect is about 8 to
`
`about 24 hours; and (c) relative oxymorphone bioavailability is in the range of about 0.5 to about
`
`1.5 compared to an orally-administered aqueous solution of oxymorphone.
`
`In one embodiment of the invention, the oxymorphone or salt of oxymorphone is
`
`dispersed in a controlled release delivery system that comprises a hydrophilic material which
`
`upon exposure to gastrointestinal fluid forms a gel matrix that releases oxymorphone at a
`
`controlled rate. The rate of release of oxymorphone from the matrix depends on the drug's
`
`partition coefficient between components of the matrix and the aqueous phase within the
`
`gastrointestinal tract. In a preferred form of this embodiment, the hydrophilic material of the
`
`controlled release delivery system comprises a mixture of a heteropolysaccharide gum and an
`
`agent capable of cross-linking the heteropolysaccharide in the presence of gastrointestinal fluid.
`
`The controlled release delivery system may also comprise a water-soluble pharmaceutical diluent
`
`mixed with the hydrophilic material. (cid:9)
`
`Preferably, the cross-linking agent is a
`
`homopolysaccharide gum and the inert pharmaceutical diluent is a mono-saccharide, a
`
`disaccharide, and polyhydric alcohol, or a mixture thereof.
`
`In a specific preferred embodiment, the oxymorphone is present as oxymorphone
`
`hydrochloride, the weight ratio of heteropolysaccharide to homopolysaccharide is in the range of
`
`about 1:3 to 3:1, the weight ratio of heteropolysaccharide to diluent is in the range of about 1:8 to
`
`8:1, and the weight ratio of heteropolysaccharide to oxymorphone hydrochloride is in the range
`
`of about 10:1 to 1:10. A preferred heteropolysaccharide is xanthan gum and a preferred
`
`homopolysaccharide is locust bean gum. The dosage form also comprises a cationic cross-
`
`4
`
`935090_1
`
`

`

`linking agent and a hydrophobic polymer. In the most preferred embodiment, the dosage form is
`
`a tablet containing about 5 mg to about 80 mg of oxymorphone hydrochloride.
`
`The invention includes a method which comprises administering once or twice a day to a
`
`patient suffering moderate to severe, acute or chronic pain (e.g., pain associated with cancer,
`
`autoimmune diseases, infections, surgical and accidental traumas) and oxymorphone CR oral
`
`solid dosage form of the invention in an amount sufficient to alleviate the pain for a period of
`
`about 12 hours to about 24 hours.
`
`The invention also includes a method of making an oxymorphone controlled release oral
`
`solid dosage form of the invention which comprises mixing particles of oxymorphone or a
`
`pharmaceutically acceptable salt of oxymorphone with granules comprising the controlled
`
`release delivery system, preferably followed by directly compressing the mixture to form tablets.
`
`DETAILED DESCRIPTION OF THE INVENTION
`
`Pharmaceutically accepted salts of oxymorphone which can be used in this invention
`
`include salts with all those inorganic and organic acids which are commonly used to produce
`
`nontoxic salts of medicinal agents containing amine functions. Illustrative examples would be
`
`those salts formed by mixing oxymorphone with hydrochloric, sulfuric, nitric, phosphoric,
`
`phosphorous, hydrobromic, maleric, malic, ascorbic, citric or tartaric, pamoic, lauric, stearic,
`
`palmitic, oleic, myristic, lauryl sulfuric, napthalinesulfonic, linoleic or linolenic acid, and the
`
`like. The hydrochloride salt is preferred.
`
`The oxymorphone CR oral solid dosage form of this invention can be made as described
`
`for CR oral solid dosage forms of other opioid analgesics in Oshlack et al. US Patent 5,226,331
`
`and Chaisin et al. US Patent 5,958,459, the entire disclosures of which are incorporated herein.
`
`In one embodiment, a core comprising oxymorphone or oxymorphone salt is coated with
`
`a CR film which comprises a water insoluble material and which upon exposure to
`
`gastrointestinal fluid releases oxymorphone from the core at a controlled rate. In a second
`
`embodiment, the oxymorphone or oxymorphone salt is dispersed in a controlled release delivery
`
`system that comprises a hydrophilic material which upon exposure to gastrointestinal fluid forms
`
`a gel matrix that releases oxymorphone at a controlled rate. A third embodiment is a
`
`5
`
`935090_1
`
`

`

`combination of the first two-a CR matrix coated with a CR film. In any of these embodiments,
`
`the dosage form can be a tablet, a plurality of granules in a capsule, or other suitable form, and
`
`can contain lubricants, colorants, diluents, and other conventional ingredients.
`
`CR coating embodiment
`
`In this embodiment, a core comprising oxymorphone or oxymorphone salt is coated with
`
`a CR film which comprises a water insoluble material. The film can be applied by spraying an
`
`aqueous dispersion of the water insoluble material onto the core. Suitable water insoluble
`
`materials include alkyl celluloses, acrylic polymers, waxes (alone or in admixture with fatty
`
`alcohols), shellac and zein. The aqueous dispersions of alkyl celluloses and acrylic polymers
`
`preferably contain a plasticizer such as triethyl citrate, dibutyl phthalate, propylene glycol, and
`
`polyethylene glycol. (cid:9)
`
`The film coat can contain a water soluble material such as
`
`polyvinylpyrrolidone (PVP) or hydroxypropylmethylcellulose (HPMC).
`
`The core can be a granule made, for example, by wet granulation of mixed powders of
`
`oxymorphone or oxymorphone salt and a binding agent such as HPMC, or by coating an inert
`
`bead with oxymorphone or oxymorphone salt and a binding agent such as HPMC, or by
`
`spheronizing mixed powders of oxymorphone or oxymorphone salt and a spheronizing agent
`
`such as microcrystalline cellulose. The core can be a tablet made by compressing such granules
`
`or by compressing a powder comprising oxymorphone or oxymorphone salt.
`
`The in vitro and in vivo release characteristics of this CR dosage form can be modified by
`
`using mixtures of different water insoluble and water soluble materials, using different
`
`plasticizers, varying the thickness of the CR film, including release-modifying agents in the
`
`coating, or by providing passageways through the coating.
`
`CR matrix embodiment
`
`In this embodiment, the oxymorphone or oxymorphone salt is dispersed in a controlled
`
`release delivery system that comprises a hydrophilic material (gelling agent) which upon
`
`exposure to gastrointestinal fluid forms a gel matrix that releases oxymorphone at a controlled
`
`rate. Such hydrophilic materials include gums, cellulose ethers, acrylic resins, and protein-
`
`derived materials. Suitable cellulose ethers include hydroxyalkyl celluloses and carboxyalkyl
`
`6
`
`935090_1
`
`

`

`celluloses, especially hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), HPMC,
`
`and carboxy methylcellulose (CMC). Suitable acrylic resins include polymers and copolymers
`
`of acrylic acid, methacrylic acid, methyl acrylate and methyl methacrylate. Suitable gums
`
`include heteropolysaccharide and homopolysaccharide gums, e.g., xanthan, tragacanth, acacia,
`
`karaya, alginates, agar, guar, hydroxypropyl guar, carrageenan, and locust bean gums.
`
`The preferred hydrophilic material comprises a heteropolysaccharide such as xanthan
`
`gum and a cross-linking agent capable of cross-linking the heteropolysaccharide, as disclosed in
`
`Baichwal et al. US Patents 4,994,276, 5,128,143 and 5,135,757, the entire disclosures of which
`
`are incorporated herein. The cross-linking agent can be a homopolysaccharide, preferably a
`
`galactomannan gum such as locust bean gum. Preferably, the ratio of heteropolysaccharide to
`
`homopolysaccharide is in the range of about 1:9 to about 9:1, preferably about 1:3 to about 3:1.
`
`In addition to the hydrophilic material, the controlled release delivery system can also
`
`contain an inert pharmaceutical diluent such as a monosaccharide, a disaccharide, a polyhydric
`
`alcohol and mixtures thereof. The ratio of diluent to hydrophilic matrix-forming material is
`
`generally in the range of about 1:3 to about 3:1.
`
`The controlled release delivery system may also contain a cationic cross-linking agent
`
`such as calcium sulfate in an amount sufficient to cross-link the gelling agent and increase the
`
`gel strength, and an inert hydrophobic material such as ethyl cellulose in an amount sufficient to
`
`slow the hydration of the hydrophilic material without disrupting it. Preferably, the controlled
`
`release delivery system is prepared as a pre-manufactured granulation. See Baichwal US Patents
`
`5,455,046, 5,554,387, and 5,512,297, the entire disclosures of which are incorporated herein.
`
`Examples 1-2
`
`EXAMPLES
`
`Two controlled release delivery systems are prepared by dry blending xanthan gum,
`
`locust bean gum, calcium sulfate dehydrate, and dextrose in a high speed mixed/granulator for 3
`
`minutes. A slurry is prepared by mixing ethyl cellulose with alcohol. While running
`
`choppers/impellers, the slurry is added to the dry blended mixture, and granulated for another 3
`
`minutes. The granulation is then dried to a LOD (loss on drying) of less than about 10% by
`
`7
`
`935090_1
`
`

`

`weight. The granulation is then milled using 20 mesh screen. The relative quantities of the
`
`ingredients are listed in the table below.
`
`Table la
`
`Controlled Release Delivery System
`
`Example 1
`
`Excipient
`
`Locust Bean Gum, FCC
`
`Xanthan Gum, NF
`
`Dextrose, USP
`
`Calcium Sulfate Dihydrate, NF
`
`Ethylcellulose, NF
`
`Alcohol, SD3A (Anhydrous)1
`
`Total
`
`25.0
`
`25.0
`
`35.0
`
`10.0
`
`5.0
`
`(10)1
`
`100.0
`
`Example 2
`
`
`30.0
`
`30.0
`
`40.0
`
`0.0
`
`0.0
`
`(20.0)-1
`
`100.0
`
`1. Volatile, removed during processing
`
`Examples 3 to 7
`
`A series of tablets containing different amounts of oxymorphone hydrochloride were
`
`prepared using the controlled release delivery system of Example 1. The quantities of
`
`ingredients per tablet are as listed in the following table.
`
`Table 4
`
`Component
`
`Oxymorphone HCI, USP
`
`Controlled release delivery system
`
`Silicified mmicrocrystalline
`cellulose, N.F.
`Sodium stearyl fumarate, NF
`Total weight
`Opadry (colored)
`Opadry (clear)
`
`Examples 8 and 9
`
`Ex. 3 (cid:9)
`
`Ex. 4 (cid:9)
`
`Ex. 5 Ex. 6 Ex. 7
`
`mg
`
`5
`
`160
`
`20
`
`2
`187
`7.48
`0.94
`
`mg
`
`10
`
`160
`
`20
`
`2
`192
`7.68
`0.96
`
`mg
`
`20
`
`160
`
`20
`
`2
`202
`8.08
`1.01
`
`mg
`
`40
`
`mg
`
`80
`
`160
`
`160
`
`20
`
`20
`
`2
`2
`222
`262
`8.88 10.48
`1.11
`1.31
`
`8
`
`935090_1
`
`

`

`Two batches of tablets were prepared as described above for Examples 1-7, using the
`
`controlled release delivery system of Example 1. One batch was formulated to provide relatively
`
`fast controlled release, the other batch was formulated to provide relatively slow controlled
`
`release. Compositions of the tablets are shown in the following table.
`
`Table 6a
`
`Ingredients
`
`Oxymorphone HCI, USP
`
`Controlled Release Delivery
`
`System
`
`Silicified Microcrystalline
`
`Cellulose, NF
`
`Sodium stearyl fumarate, NF
`
`Coating (color)
`
`Total weight
`
`Example
`
`slow release
`
`mg/tab
`
`Example
`
`fast release
`
`mg/tab
`
`20
`
`360
`
`20
`
`4
`
`12.12
`
`416.12
`
`20
`
`160
`
`20
`
`2
`
`12.12
`
`214.12
`
`The tables of Examples 8 and 9 were tested for in vitro release rate according to USP
`
`Procedure Drug Release USP 24. Results are shown in the following table.
`
`Table 7a
`
`Time (hr)
`
`0.5
`
`1
`
`2
`
`3
`
`4
`
`5
`
`6
`
`8
`
`Example 8
`
`Example 9
`
`slow release
`
`fast release
`
`18.8
`
`27.8
`
`40.5
`
`50.2
`
`58.1
`
`64.7
`
`70.2
`
`79.0
`
`21.3
`
`32.3
`
`47.4
`
`58.5
`
`66.9
`
`73.5
`
`78.6
`
`86.0
`
`9
`
`935090_1
`
`

`

`10
`
`12
`
`85.3
`
`89.8
`
`90.6
`
`93.4
`
`Example 10: Clinical Study
`
`A clinical study was conducted to (1) assess the relative bioavailability (rate and extent of
`
`absorption) of oxymorphone CR (20 mg) (fast release formulation of Example 9) compared to
`
`oral solution oxymorphone (10 mg) under fasted conditions, (2) to assess the relative
`
`bioavailability of oxymorphone CR (20 mg) compared to oral solution oxymorphone (10 mg)
`
`under fed conditions, (3) to assess the relative bioavailability of oxymorphone CR (20 mg) fed
`
`compared to oxymorphone CR (20 mg) fasted, (4) to assess the relative bioavailability of oral
`
`solution oxymorphone fed compared to oral solution oxymorphone fasted, and (5) to assess the
`
`relative safety and tolerability of controlled-release oxymorphone (20 mg) under fed and fasted
`
`conditions.
`
`Study Design and Conduct
`
`This study had a single-center, open-label, analytically blinded, randomized, four-way
`
`crossover design. Subjects randomized to Treatment A and Treatment C, as described below,
`
`were in a fasted state following a 10-hour overnight fast. Subjects randomized to Treatment B
`
`and Treatment D, as described below, were in the fed state, having had a high fat meal,
`
`completed ten minutes prior to dosing. There was a 14-day washout interval between the four
`
`dose administrations. The subjects were confined to the clinic during each study period. Subject
`
`assigned to receive Treatment A and Treatment B were discharged from the clinic on Day 3
`
`following the 48-hour procedures, and subjects assigned to receive Treatment C and Treatment D
`
`were discharged from the clinic on Day 2 following the 36-hour procedures. On Day 1 of each
`
`study period the subjects received one of four treatments:
`
`Treatments A and B: oxymorphone CR 20 mg tablets. Subjects randomized to
`
`Treatment A received a single oral dose of one 20 mg oxymorphone CR tablet taken with 240
`
`mL of water after a 10-hour fasting period. Subjects randomized to Treatment B received a
`
`single oral dose of one 20 mg oxymorphone CR tablet taken with 240 mL of water 10 minutes
`
`after a standardized high fat meal.
`
`10
`
`935090_1
`
`(cid:9)
`

`

`Treatments C and D: oxymorphone HC1 solution, USP, 1.5 mg/mL injection 10 mL
`
`vials. Subjects randomized to Treatment C received a single oral dose of 10 mg (6.7 mL)
`
`oxymorphone solution taken with 240 mL of water after a 10-hour fasting period. Subjects
`
`randomized to Treatment D received a single oral dose of 10 mg (6.7mL) oxymorphone solution
`
`taken with 240 mL of water 10 minutes after a standardized high-fat meal.
`
`A total of 28 male subjects were enrolled in the study, and 24 subjects completed the
`
`study. The mean age of the subjects was 27 years (range of 19 through 38 years), the mean
`
`height of the subjects was 69.6 inches (range of 64.0 through 75.0 inches), and the mean weight
`
`of the subjects was 169.0 pounds (range 117.0 through 202.0 pounds). The subjects were not to
`
`consume any alcohol-, caffeine-, or xanthine-containing foods or beverages for 24 hours prior to
`
`receiving study medication for each study period. Subjects were to be nicotine and tobacco free
`
`for at least 6 months prior to enrolling in the study. In addition, over-the-counter medications
`
`were prohibited 7 days prior to dosing and during the study. Prescription medications were not
`
`allowed 14 days prior to dosing and during the study.
`
`The subjects were screened within 14 days prior to study enrollment. The screening
`
`procedure included medical history, physical examination (height, weight, frame size, vital signs,
`
`and ECG), and clinical laboratory tests (hematology, serum chemistry, urinalysis, HIV antibody
`
`screen, Hepatitis B surface antigen screen, Hepatitis C antibody screen, and a screen for
`
`cannabinoids).
`
`During the study, the subjects were to remain in an upright position (sitting or standing)
`
`for 4 hours after the study drug was administered. Water was restricted 2 hours predose to 2
`
`hours postdose. During the study, the subjects were not allowed to engage in any strenuous
`
`activity.
`
`Subjects reported to the clinic on the evening prior to each dosing. The subjects then
`
`observed a 10-hour overnight fast. On Day 1, subjects randomized to Treatment B and
`
`Treatment D received a high-fat breakfast within 30 minutes prior to dosing. A standardized
`
`meal schedule was then initiated with lunch 4 hours postdose, dinner 10 hours postdose, and a
`
`snack 13 hours postdose. On Day 2, a standardized meal was initiated with breakfast at 0815,
`
`11
`
`935090_1
`
`

`

`lunch at 1200, and dinner at 1800. Subjects randomized to Treatment A and Treatment B
`
`received a snack at 2100 on Day 2.
`
`Vital signs (sitting for 5 minutes and consisting of blood pressure, pulse, respiration, and
`
`temperature), and 12-lead ECG were assessed at the —13 hour of each check-in period and at the
`
`completion of each period. A clinical laboratory evaluation (hematology, serum chemistry,
`
`urinalysis) and a brief physical examination were performed at the —13 hour of each check-in
`
`period and at the completion of the each period. Subjects were instructed to inform the study
`
`physician and/or nurses of any adverse events that occurred during the study.
`
`Blood samples (7 mL) were collected during each study period at the 0 hour (predose),
`
`and at 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 14, 16, 20, 24, 30, 36, and 48 hours post-dose (19
`
`samples) for subjects randomized to Treatment A and Treatment B. Blood samples (7 mL) were
`
`collected during each study period at the 0 hour (predose), and at 0.25, 0.5, 0.75, 1, 1.25, 1.5,
`
`1.75, 2, 3, 4, 5, 6, 8, 10, 12, 14, 16, 20, and 36 hours post-dose (21 samples) for subjects
`
`randomized to Treatment C and Treatment D. A total of 80 blood samples (560 mL) per subject
`
`were drawn during the study for drug analysis. Plasma samples were separated by
`
`centrifugation, and then frozen at —70°C, and kept frozen until assayed.
`
`Analytical Method
`
`An LC/MS/MS method was developed and validated for the determination of
`
`oxymorphone in human EDTA plasma. Samples were spiked with internal standard, d3_
`oxymorphone, and placed on the RapidTrace® for automatic solid phase extraction. Extracts
`
`were dried under nitrogen and reconstituted with acetonitrile before injection onto an
`
`LC/MS/MS. The Perkin Elmer Sciex API III+, or equivalent using a turbo ion spray interface,
`
`was employed in this study. Positive ions were monitored in the MRM mode.
`
`Pharmacokinetic and Statistical Methods
`
`The following pharmacokinetic parameters were computed from the plasma
`
`oxymorphone concentration-time data:
`
`12
`
`935090_1
`
`

`

`AUC(0-t)
`
`AUC(0-inf)
`
`AUC(0-24)
`
`Cmax
`Tmax
`Kel
`
`T1/2e1
`
`Area under the drug concentration-time curve from time zero to the time of
`the last quantifiable concentration (Ct), calculated using linear trapezoidal
`summnation.
`Area under the drug concentration-time curve from time zero to infinity.
`AUC(0-inf) = AUC(0-t) + Ct/Kel, where Kel is the terminal elimination
`rate constant.
`Partial area under the drug concentration-time curve from time zero to 24
`hours.
`Maximum observed drug concentration.
`Time of the observed maximum drug concentration.
`Elimination rate constant based on the linear regression of the terminal
`linear portion of the LN(concentration) time curve.
`Half life, the time required for the concentration to decline by 50%,
`calculated as LN(2)/Kel
`
`Terminal elimination rate constants were computed using linear regression of a minimum
`
`of three time points, at least two of which were consecutive. Kel values for which correlation
`
`coefficients were less than or equal to 0.8 were not reported in the pharmacokinetic parameter
`
`tables or included in the statistical analysis. Thus, T1/2e1, AUC(0-inf), Cl/F, MRT, and LN-
`
`transformed T1/2e1, AUC(0-inf), and Cl/F were also not reported in these cases.
`
`A parametric (normal-theory) general linear model was applied to each of the above
`
`parameters (excluding Tmax and Frel), and the LN-transformed parameters Cmax, AUC(0-24),
`
`AUC(0-t), AUC(0-inf), Cl/F, and Tl/2el. Initially, the analysis of variance (ANOVA) model
`
`included the following factors: treatment, sequence, subject within sequence, period, and
`
`carryover effect. If carryover effect was not significant, it was dropped from the model. The
`
`sequence effect was tested using the subject within sequence mean square, and all other main
`
`effects were tested using the residual error (error mean square). The following treatment
`
`comparisons of relative rate and extent of absorption were made: Treatment B versus Treatment
`
`A, Treatment A versus Treatment C (dose normalized to 20 mg). Treatment B versus Treatment
`
`D (dose normalized to 20 mg), and Treatment D versus Treatment C (dose normalized to 20 mg
`
`for both treatments). The 90% confidence intervals of the ratios of the treatment least squares
`
`parameter means were calculated. Tmax was analyzed using the Wilcoxon Signed Ranks test.
`
`Summary statistics were presented for Frel.
`
`13
`
`935090_1
`
`

`

`Plasma oxymorphone concentrations were listed by subject at each collection time and
`
`summarized using descriptive statistics. Pharmacokinetic parameters were also listed by subject
`
`and summarized using descriptive statistics.
`
`Results
`
`A total of 26 analytical runs were required to process the clinical samples from this study.
`
`Of these 26 analytical runs, 26 were acceptable for oxymorphone. Standard curves for the 26
`
`analkytical runs in EDTA plasma used in this study covered a range of 0.0500 to 20.000 mg/mL
`
`with a limit of quantitation of 0.0500 ng/mL for both compounds. Quality control samples
`
`analyzed with each analytical run had coefficients of variation less than or equal to 14.23% for
`
`oxymorphone.
`
`A total of 28 subjects received at least one treatment. Only subjects who completed all 4
`
`treatments were included in the summary statistics and statistical analysis.
`
`The mean oxymorphone plasma concentration versus time curves for Treatments A, B, C,
`
`and D are presented in Figure 1 (linear scale, with S.D.). Figure 2 (linear scale, without S.D.)
`
`and Figure 3 (semi-log scale).
`
`Individual concentration versus time curves were characterized by multiple peaks which
`
`occurred in the initial 12-hour period following the dose. In addition, a small "bump" in plasma
`
`oxymorphone concentration was generally observed in the 24 to 48 hour post-dose period.
`
`The arithmetic means of the plasma oxymorphone pharmacokinetic parameters and the
`
`statistical for Treatment B versus Treatment A are summarized in table 1.
`
`TABLE 1
`
`Summary of the Pharmacokinetic Parameters of Plasma Oxymorphone for Treatments B and A
`Plasma Oxymorphone (cid:9)
`
`Treatment A (cid:9)
`Treatment B
`
`Pharmacokinetic (cid:9)
`Parameters (cid:9)
`Cmax(ng/mL) (cid:9)
`
`Tmax(hr) (cid:9)
`
`Arithmetic (cid:9)
`Arithmetic (cid:9)
`Mean
`Mean (cid:9)
`Mean (cid:9)
`90% CI (cid:9)
`SD (cid:9)
`SD (cid:9)
`Ratio
`1.7895 0.6531 1.1410 0.4537 125.4-191.0 158.2
`
`5.65 (cid:9)
`
`9.39 (cid:9)
`
`5.57 (cid:9)
`
`7.14
`
`14
`
`935090_1
`
`(cid:9)
`

`

`Auc(0-24)(ng*hr/mL)
`
`AUC(0-0(ng*hr/mL)
`
`AUC(0-inf)(ng*hr/mL)
`
`T 1/2e1(hr)
`
`14.27
`
`19.89
`
`21.29
`
`12.0
`
`4.976
`
`6.408
`
`6.559
`
`3.64
`
`11.64
`
`17.71
`
`19.29
`
`12.3
`
`3.869
`
`8.471
`
`5.028
`
`3.99
`
`110.7-134.0
`
`122.3
`
`100.2-123.6
`
`111.9
`
`105.3-133.9
`
`119.6
`
`57.4-155.2
`
`106.3
`
`Treatment B - 1 x 20 mg oxymorphone CR Tablet, Fed: test
`
`Treatment A = 1 x 20 mC oxymorphone CR Tablet, Fasted: reference
`
`The arithmetic means of the plasma oxymorphone pharmacokinetic parameters and the
`
`statistical comparisons for Treatment A versus Treatment C are summarized in table 2.
`
`TABLE 2
`
`Summary of the Pharmacokinetic Parameters of Plasma Oxymorphone for Treatments A and C
`
`Plasma Oxymorphone (cid:9)
`
`
`
`Treatment A
`
`Treatment C
`
`SD
`
`Arithmetic
`Mean
`2.2635
`0.978
`12.39
`14.53
`18.70
`16.2
`
`SD
`1.0008
`1.14
`4.116
`4.909
`6.618
`11.4
`
`90% CI
`33.4-66.0
`
`82.8-104.6
`107.7-136.3
`80.2-108.4
`32.9-102.1
`
`Mean
`Ratio
`49.7
`
`93.7
`122.0
`94.3
`67.5
`
`Arithmetic
`Pharmacokinetic
`Parameters
`Mean
`0.4537
`1.1410
`Cmax(ng/mL)
`7.14
`5.57
`Tmax(hr)
`11.64
`Auc(0-24)(ng*hr/mL)
`3.869
`8.471
`17.71
`AUC(0-I)(ng*hr/mL
`19.29
`AUC(0-inf)(ng*hr/mL)
`5.028
`3.99
`12.3
`T 1/2e1(hr)
`Treatment A = 1 x 20 mg oxymorphone CR Tablet, Fasted: test
`Treatment C = 10 mg/6.7 mL oxymorphone HCI Oral Solution, Fasted: Dose Normalized to 20 ng: reference.
`
`The arithmetic means of the plasma oxymorphone pharmacokinetic parameters and the
`
`statistical comparisons for Treatment D versus Treatment C are summarized in table 3.
`
`TABLE 3
`
`Summary of the Pharmacokinetic Parameters of Plasma Oxymorphone for Treatments A and C
`Plasma Oxymorphone (cid:9)
`
`Treatment B
`Treatment D
`Arithmetic
`Arithmetic
`Mean
`SD
`Mean
`1.7895
`3.2733 1.3169
`
`Pharmacokinetic
`P

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket