throbber
.
`
`I
`
`CSE
`04122/04
`
`4000.3010 us
`
`- 1 -
`
`Date: ApA_; J; 447 c2oot.f
`
`Express Mail Label No.
`
`EV 373377224 US
`
`Inventor:
`Attorney's Docket No.:
`
`Elliot Ehrich
`4000.3010 us
`
`NAL TREXONE LONG ACTING FORMULATIONS AND METHODS OF USE
`
`5
`
`10
`
`15
`
`20
`
`BACKGROUND OF THE INVENTION
`Alcohol dependence is a chronic disorder that results from a variety of genetic,
`psychological and environmental factors. Traditional treatment has consisted of two
`phases: detoxification and rehabilitation. Detoxification ameliorates the symptoms and
`signs of withdrawal; rehabilitation helps the patient avoid future problems with alcohol.
`In the past, most rehabilitative treatments have been psychosocial. With advances in
`neurobiology, there is increasing interest in drug therapy for alcohol dependence. For a
`discussion of the development of this field, see Swift, R., Drug Therapy for Alcohol
`Dependence, NEJM, May 13, 1999, 1482-1490. Yet, the successful treatment of
`alcoholism has many serious challenges and complications. Patient compliance is a
`serious problem.
`Accordingly, there is a need for improving naltrexone therapies.
`
`SUMMARY OF THE INVENTION
`The inventions described herein arose from unexpected discoveries made during
`clinical trials with a long acting formulation of naltrexone. As such, the invention
`includes a method for treating an individual in need of naltrexone comprising the step of
`parenterally administering a long acting formulation comprising naltrexone to the
`individual wherein the serum AUC of naltrexone is at least about two times, preferably
`at least about three times, more preferably about 3.3 times greater than that achieved by
`50 mg/day oral administration.
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 2 -
`
`The inventions also include a method of treating an individual in need of
`naltrexone comprising administering a long acting formulation in a dose comprising at
`least about 160 mg of naltrexone, preferably between about 160 mg and about 480 mg
`naltrexone, more preferably between about 160 and 240 mg of naltrexone or about 310
`to about 480 mg of naltrexone.
`The inventions also include a method of treating an individual in need of
`naltrexone comprising administering naltrexone, such as in a long acting formulation, in
`the absence of co-administering alcohol, to an individual who has not abstained from
`alcohol within three days, such as five days, prior to the naltrexone administration.
`The inventions include a method of increasing the days prior to occurrence of
`alcohol consumption in an individual in need of naltrexone comprising administering a
`long acting formulation comprising naltrexone, in the absence of co-administering
`alcohol, to an individual who has not abstained from alcohol within three days, such as
`five days, prior to the naltrexone administration.
`The inventions include a method of treating an individual in need of naltrexone
`comprising administering a long acting formulation comprising naltrexone in a dosage
`between about 160 mg to about 480 mg naltrexone every four weeks for a period of
`about 24 weeks or more wherein the individual has not used oral naltrexone within five
`days, such as within ten days, before said administration.
`
`BRIEF SUMMARY OF THE ORA WINGS
`Figure lA-lC shows the cumulative mean event rate of heavy drinking during
`the study by treatment group and gender. As an example, at day 100, the mean number
`of cumulative heavy drinking days for the overall .study population was 22.3 for the
`long-acting naltrexone 380 mg patients, 27.3 for long-acting naltrexone 190 mg
`patients, and 30.0 for placebo patients.
`Figure 2 shows median heavy drinking days per month for each treatment group,
`overall and by gender.
`
`5
`
`10
`
`15
`
`20
`
`25
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 3 -
`
`10
`
`15
`
`DETAILED DESCRIPTION OF THE INVENTION
`The inventions relate to the administration of a naltrexone containing
`formulation, preferably a long acting naltrexone formulation, to patients in need thereof
`and to the use of naltrexone in the manufacture of medicaments for use in such
`5 methods.
`In one embodiment, the invention includes a method for treating an individual in
`need of naltrexone comprising the step of parenterally administering a long acting
`formulation comprising naltrexone to the individual wherein the serum AUC of
`naltrexone is at least about two times, preferably at least about three times, more
`preferably about 3.3 times greater than that achieved by 50 mg/day oral administration.
`This invention arose from the unexpected discovery that substantially improved serum
`levels of naltrexone can be achieved by administering long acting formulations of
`naltrexone, such as the Alkermes, Inc. formulation, Vivitrex® injectable suspension,
`made employing its Medisorb® delivery system. Indeed, it was not expected that serum
`levels of about 3.3 times that achieved by a 50 mg/day oral dose could be achieved by a
`single IM administration of Vivitrex®.
`The inventions also include a method of treating an individual in need of
`naltrexone comprising administering naltrexone, such as in a long acting formulation, in
`the absence of co-administering alcohol, to an individual who has not abstained from
`alcohol within three days, such as five days, prior to the naltrexone administration. In
`this embodiment, it was unexpectedly discovered that good to excellent results could be
`achieved without either requiring alcohol abstinence or requiring alcohol consumption
`during therapy, as taught by Sinclair, United States Patent No. 4,882,335. Further, good
`to excellent results were achieved in patients that did not receive oral. naltrexone in
`advance of the long acting formulation administration, contrary to the clinical protocols
`as taught by Drug Abuse Sciences. Thus, the inventions also include administering a
`long acting formulation to individuals who did not receive a prior oral dose of
`naltrexone, for example, within 3, such as within about 5 days or about 10 days of
`commencing therapy.
`
`20
`
`25
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`-4-
`
`As such, the inventions also include a method of treating an individual in need
`of naltrexone comprising administering a long acting formulation in a dose comprising
`between about 160 mg and 240 mg of naltrexone or about 310 mg to about 480 mg of
`naltrexone and formulations for use in the methods described herein .. Preferred
`formulations are administered in a dose comprising about 190 mg or about 380 mg
`naltrexone.
`The naltrexone can be in any form, including anhydrous, hydrate, solvate or salt
`forms or combinations thereof. It can be crystalline or non-crystalline or combinations
`thereof. A preferred naltrexone form comprises a naltrexone ethanolate, such as that
`described in United States Patent Application No. 60/475,863, filed on June 4, 2003,
`which is incorporated herein by reference and/or anhydrous naltrexone. A particularly
`preferred naltrexone form is that produced by the encapsulation process described in
`United States Patent No. 6,264,987, by Wright et al., which is incorporated herein by
`reference.
`The naltrexone can be combined with any of the well-known biodegradable and
`bioerodible carriers, such as polylactides, poly(lactic acids) and poly-lactide-co-
`glycolides and collagen formulations. A particularly preferred polymer is a polylactide-
`co-glycolide polymer which possesses a molecular weight of at least 100,000 daltons,
`such as those described below in the exemplification. Such materials may be in the
`form of solid implants, sponges, and the like.
`As stated above, the naltrexone is preferably in a long acting formulation. Long
`acting (also referred to as extended, sustained, or controlled release) preparations may
`be achieved through the use of polymers (preferably poly-lactide or poly-lactide-co-
`glycolide polymers) to entrap or encapsulate the naltrexone described herein. Extended
`release formulations can be made by spray drying polymer-drug mixtures, emulsion-
`based technologies, coacervation based technologies, film casting, extrusion based
`technologies and other processes to manufacture polymer-drug microparticles
`possessing an extended release profile. Examples of suitable extended release
`technologies that can be used to incorporate the novel naltrexone forms described herein
`include, without limitation, the MEDISORB® technology, as described in, for example,
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 5 -
`
`10
`
`15
`
`US Patent Nos. 6,264,987 to Wright, 5,654,008 and/or 5,792,477, for example; the
`PROLEASE® technology, as described, for example in US Patent 6,358,443 to Herbert;
`the technologies described by Southern Research Institute, as described for example in
`US Patents 6,306,425 and 5,407,609; and "Method of Preparing Sustained Release
`5 Microparticles," U.S. Application No. 60/441,946, filed January 23, 2003, and the
`technologies described by Alza Corp., including the ALZAMER® Depot injection
`technology. The contents of these patents are incorporated herein by reference in their
`entirety.
`In a preferred embodiment, the long acting formulation delivers therapeutically
`beneficial amounts of naltrexone to the patient for a period of at least one week,
`preferably at least about two weeks, more preferably at least about 3 or about 4 or more
`weeks. A four week delivery is often referred to as a monthly delivery.
`In one preferred embodiment, the naltrexone is present in the extended release
`device or formulation in an amount of at least about 5% by weight, preferably at least
`about I 0% by weight, more preferably at least about 30% by weight, such as about 35%
`by weight naltrexone of the total weight of the device, or formulation.
`Alternatively, instead of incorporating naltrexone into polymeric particles, it is
`possible to entrap these materials in microparticles prepared, for example, by
`coacervation techniques or by interfacial polymerization (for ex~mple,
`hydroxymethylcellulose or gelatine-microcapsules and poly-(methylmethacrylate)
`microcapsules, respectively), in colloidal drug delivery systems (for example,
`liposomes, albumin, microparticles, microemulsions, nanoparticles, and nanocapsules),
`or in macroemulsions.
`When the composition is to be used as an injectable material, including but not
`limited to needle-less injection, it can be formulated into a conventional injectable
`carrier. Suitable carriers include biocompatible and pharmaceutically_ acceptable
`solutions. The injection can be intramuscular or subcutaneous.
`While the formulation may contain additional excipients, as is well known in the
`art, the present invention can achieve an excellen~ release profile with the simple
`formulation described herein. Such additional excipients can increase or decrease the
`
`20
`
`25
`
`30
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 6 -
`
`rate of release of the agent. Ingredients which can substantially increase the rate of
`release include pore forming agents and excipients which facilitate polymer
`degradation. For example, the rate of polymer hydrolysis is increased in non-neutral pH.
`Therefore, an acidic or a basic excipient such as an inorganic acid or inorganic base can
`be added to the polymer solution, used to form the microparticles, to alter the polymer
`erosion rate.
`A preferred embodiment of the described sustained release formulations consists
`essentially of the biocompatible polymer and naltrexone. By "consists essentially of' is
`meant the absence of ingredients which substantially increase the rate of release of the
`active agent from the formulation. Examples of additional excipients which would not
`be expected to substantially increase or decrease the rate of release of the agent include
`additional active agents and inert ingredients.
`In yet another embodiment, the formulation consists of the biocompatible
`polymer and naltrexone. By "consists of' is meant the absence of components or
`ingredients other than those listed and residual levels of starting materials, solvents, etc.
`from the process.
`As stated above, the formulation preferably releases naltrexone over a period of
`at least about one, two, three or four weeks. As such, the formulation can be
`administered using a dosing schedule which achieves the desired therapeutic levels for
`the desired period oftime. For example, the formulation can be administered and,
`optionally, the patient monitored until levels of the drug being delivered return to
`baseline. Following an actual or projected return to baseline, the formulation can be
`administered again. Alternatively, the subsequent administration of the formulation can
`occur prior to achieving baseline levels in the patient. As such, the formulation can be
`advantageously administered weekly, with a one week release formulation, biweekly.
`with a two week release formulation, or monthly with a four week release formulation.
`Vivitrex is a four week release formulation with a monthly (e.g., every four weeks)
`administration. The therapy can end after a single dose or can be maintained for longer
`periods oftime. In one embodiment, the therapy can maintained for at least about 4, 8,
`12, 16, 20 and 24 weeks or more. Where more than one administration is given, the
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 7 -
`
`5
`
`10
`
`15
`
`20
`
`25
`
`second administration can be given at least about 7 days, preferably at least about 14
`days, more preferably at least about 21 days, such as about 28 days, after the first
`administration and combinations thereof. In this context "about" preferably means
`within three days of the targeted date.
`Particularly good results were achieved upon administering the same
`formulation and same dose with each administration. Thus, where a 380 mg dose was
`given in the first administration, good to excellent results were achieved when the
`second and subsequent doses were about 380 mg. Surprisingly, good to excellent
`results were also obtained when each dose administered contained 190 mg naltrexone,
`particularly in men and women who were abstinent from alcohol in the three or five
`days prior to commencing treatment. As such, in one embodiment of the invention, the
`doses and/or formulations administered in each subsequent administration were similar
`or the same. As stated above, the formulation is preferably a microsphere formulation
`and is administered by IM injection. Administration to the buttock in a volume of up to
`about 4 mL in an injectable diluent was performed in the trials leading up to these
`inventions.
`The methods of the inventions achieved good to excellent results in women and,
`in particular, men. Good to excellent results were achieved in young individuals
`(defined as less than 50 years of age), particularly men. Individuals afflicted by alcohol
`dependency, such as a heavy drinker achieved good to excellent results. A heavy
`drinker is understood in the art to include women who consume four or more alcoholic
`beverages in a day and men who consume five or more alcoholic beverages in a day.
`In yet another embodiment, the inventions include a method of increasing the
`days prior to occurrence of alcohol consumption in an individual in need of naltrexone
`comprising administering a long acting formulation comprising naltrexone in
`accordance with the protocols and/or dosing regimens described herein. In one
`embodiment, the increase in days prior to occurrence of alcohol consumption can
`include the consumption of a single alcoholic beverage or it can include consumption of
`four or five alcoholic beverages, such as the number of drinks characterizing an episode
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 8 -
`
`of "heavy drinking," above. In general, the greater the number of days that transpire
`prior to alcohol consumption indicates a more successful therapy.
`
`EXEMPLIFICATION
`5 Method for Manufacturing Vivitrex® Long Acting Formulations
`
`The invention includes a preferred method for manufacturing ·extended release
`devices, wherein the resulting device contains a mixture of the described polymorphic
`forms.
`
`Polymer solution can be formed by dissolving a poly(lactide )-co-glycolide
`polymer, such as a 75:25 DL PLGA (poly(lactide)-co-glycolide) in a polymer solvent,
`such as ethyl acetate (EtAc), to form a solution. Preferred PLGA polymers are high
`molecular weight polymers, such as polymers possessing a molecular weight of at least
`about 100,000 daltons. A naltrexone solution can be formed by dissolving naltrexone
`base in a suitable solvent, such as benzyl alcohol (BA), to form a solution. The polymer
`solution and the naltrexone solution are preferably mixed together to form a
`drug/polymer solution that will be the "organic" or "oil" phase of the. emulsion.
`The "aqueous" or "continuous" phase of the emulsion (emulsifying solution) is
`prepared. The aqueous phase preferably contains poly(vinyl alcohol) (PVA) and
`polymer solvent, such .as EtAc. The organic phase and the aqueous phase can be
`conveniently combined in a first static mixer to form an oil-in-water emulsion.
`In an optional partial extraction step, the emulsion flows out of the first static
`mixer and into a second static mixer where the emulsion can be combined with a
`primary extraction solution which enters the second static mixer. The primary
`extraction solution (such as can be formed by an EtAc aqueous solution) can initiate
`solvent extraction from the microdroplets of the emulsion during the partial primary
`extraction step in the second static mixer.
`The outflow of the first or second static mixer can flow into ah extraction vessel
`containing primary extraction solution. The solvents (BA and EtAc) are substantially
`extracted from the organic phase of the emulsion in this primary solvent extraction step,
`
`10
`
`15
`
`20
`
`25
`
`30
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 9 -
`
`5
`
`10
`
`15
`
`20
`
`· 25
`
`resulting in nascent microparticles comprised mainly of polymer and drug. The primary
`solvent extraction step lasts for approximately six hours.
`The microparticles can be collected, and vacuum dried, optionally with a
`nitrogen bleed using a customized vibratory sieve. After collection and prior to drying,
`the microparticles are rinsed with a 25% ethanol solution that removes the emulsifying
`agent (PVA), and enhances yield by aiding in the transfer of the microparticles to the
`cold dryer. This step is conducted, preferably at cold temperatures, until the desired
`level of dryness is achieved. As can be seen in the examples below, the degree of
`dryness (as measured, for example, by a humidity probe), impacts upon the degree of
`crystallinity achieved in the final product. For example, it can be advantageous to select
`a drying time of at least about 8, 16, 24 or 40 hours of drying. For example, it can be
`advantageous to select a drying time of at least about 8, 16, 24 or 40 hours where drying
`is 40%, 70%, 95% or 100% complete respectively. Drying is considered complete when
`the absolute humidity of the effluent gas reaches approximately 0 g/m3•
`The microparticles can then be resuspended in a second extraction solution. The
`second solution can contain the solvent desired to form the polymorphic form, such as
`ethanol. For example, a solution comprising at least about 10% by volume, preferably
`at least about 20% ethanol, can be used. This can be conveniently called the reslurry
`and secondary solvent extraction steps. The solvent, such as ethanol, can facilitate
`further extraction of BA and EtAc. Further, the crystallinity of the drug increases
`during the step. The secondary solvent extraction step is carried out in an extraction
`vessel for approximately two, three, four or more hours. This step can be conveniently
`completed at room temperature. However, other temperatures can be selected as well.
`In the collection/final dry step, the microparticles are collected, and vacuum dried with
`a nitrogen bleed using a customized vibratory sieve.
`In the final harvest step, the microparticles can be transferred into a sterile
`container and stored, for example, in a freezer at -20°C, until filling into vials.
`Preferably, the stored microparticles are sieved through a 150 micro.n screen to remove
`any oversized material prior to filling into vials.
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 10 -
`
`Example 1
`The naltrexone base microparticles were produced using a co~solvent extraction
`process. The theoretical batch size was 15 to 20 grams. The polymer (MEDISORB®
`7525 DL polymer, MEDISORB® 8515 DL polymer and MEDISORB® 6536 DL
`polymer, all available from Alkermes, Inc., Blue Ash, Ohio.) was dissolved in ethyl
`acetate to produce a 16. 7% w/w polymer solution. The naltrexone base anhydrous was
`dissolved in benzyl alcohol to produce a 30.0% w/w solution. In various batches, the
`amount of drug and polymer used was varied to produce microparticles with different
`theoretical drug loading ranging from 30%-75%. The ambient polymer and drug
`solutions were mixed together until a single homogeneous solution (organic phase) was
`produced. The aqueous phase was at ambient conditions and contained I% w/w
`polyvinyl alcohol and a saturating amount of ethyl acetate. These two solutions were
`pumped via positive displacement pumps at a ratio of 3: 1 (aqueous: organic) through a
`114" in-line mixer to form an emulsion. The emulsion was transferred to a stirring
`solvent extraction solution consisting of 2.5% w/w of ethyl acetate dissolved in distilled
`water at 5-10° C, at a volume of 0.5L of extraction solution per theoretical gram of
`microparticles. Both the polymer and drug solvents were extracted into the extraction
`solution from the emulsion droplets to produce microparticles. The initial extraction
`process ranged from two to four hours. The microparticles were collected on a 25 µm
`sieve and rinsed with a cold ( <5°C) 25% w/w ethanol solution. The microparticles were
`dried cold overnight (approximately 17 hours) using nitrogen. The microparticles were
`then transferred to the reslurry solution, which consisted of a vigorously stirring 25%
`w/w ethanol solution at 5-10°C After a short mixing time (five to fifteen minutes), the
`reslurry solution and the microparticles were transferred to a stirring 25% w/w ethanol
`~econdary extraction solution (approximately 25°C at a volume of0.2 L of secondary
`extraction solution per theoretical gram of microparticles ). The microparticles stirred
`for six hours enabling additional solvent removal from the microparticles to take place.
`The microparticles were then collected on a 25 µm sieve and rinsed 'Yith a 25% w/w
`ethanol solution at ambient temperature. These microparticles dried in a hood under
`ambient conditions overnight (approximately 17 hours), were sieved to remove
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 11 -
`
`agglomerated microparticles and then placed into a freezer for storage.
`
`5
`
`10
`
`15
`
`20
`
`25
`
`Example 2
`A 1 kg batch of naltrexone microspheres was prepared as follows. Polymer
`solution was formed by dissolving 75:25 DL PLGA (poly(lactide)-co-glycolide) in ethyl
`acetate (EtAc) to form a solution of 16.7% polymer and 83.3% EtAc. A naltrexone
`solution was formed by dissolving naltrexone base in benzyl alcohol (BA) to form a
`solution of 30% naltrexone base anhydrous and 70% BA. The polyrrier solution and the
`naltrexone solution were mixed together to form a drug/polymer solution that was the
`"organic" or "oil" phase of the emulsion.
`The "aqueous" or "continuous" phase of the emulsion (emulsifying solution)
`was prepared by dissolving poly( vinyl alcohol) (PV A) and EtAc in water-for-injection
`(WFI). The organic phase and the aqueous phase were combined in a first static mixer
`to form an oil-in-water emulsion. The droplet size of the emulsion was determined by
`controlling the flow rates of the two phases through the first static mixer.
`In a partial primary extraction step, the emulsion flowed out of the first static
`mixer and into a second static mixer where the emulsion was combined with a Primary
`extraction solution which enters the second static mixer. The primary extraction
`solution (2.5% EtAc and 97 .5% WFI at approximately 6°C) initiated solvent extraction
`from the microdroplets of the emulsion during the partial primary extraction step in the
`second static mixer.
`The outflow of the second static mixer (combined flow stream of the emulsion
`and the primary extraction solution) flowed into an extraction vessel containing primary
`extraction solution. The solvents (BA and EtAc) were further extracted from the
`organic phase of the emulsion in this primary solvent extraction step, resulting in
`nascent microparticles comprised mainly of polymer and drug. The primary solvent
`extraction step lasted for approximately six hours.
`The microparticles were collected, and vacuum dried with a nitrogen bleed using
`a customized vibratory sieve. After collection and prior to drying, the microparticles
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 12 -
`
`5
`
`10
`
`15
`
`20
`
`25
`
`were rinsed with a 25% ethanol solution that removes the emulsifying agent (PV A), and
`enhances yield by aiding in the transfer of the microparticles to the dryer.
`To further reduce the solvent levels the microparticles were resuspended in a
`second extraction solution of 25% ethanol and 75% WFI in the reslurry and secondary
`solvent extraction steps. The ethanol facilitated further extraction of BA and EtAc. The
`secondary solvent extraction step was carried out in an extraction vessel for
`approximately four hours. In the collection/final dry step, the microparticles were
`collected, and vacuum dried with a nitrogen bleed using a second customized vibratory
`sieve.
`
`In the final harvest step, the microparticles were transferred into a sterile
`container and stored in a freezer at -20°C until filling into vials. Preferably, the stored
`microparticles were sieved through a 150 micron screen to remove any oversized
`material prior to filling into vials.
`
`Example 3
`Screening and Eligibility Criteria
`Participants were male or nonpregnant, nonlactating female outpatients ~ 18
`years old with a current diagnosis of alcohol dependence. Patients had a minimum of
`two episodes of heavy drinking (~5 alcoholic drinks/day for men and.~4 drinks/day for
`women) per week during the 30 days prior to screening.
`Exclusion criteria included evidence of liver failure; alanine aminotransferase
`(ALT) or aspartate aminotransferase (AST) levels greater than three times the upper
`limit of normal; history of pancreatitis; major depression with suicidal ideation,
`psychosis, or bipolar disorder (patients with treated depression and stable
`pharmacotherapy for at least 8 weeks were not excluded); dependence within the past
`year on benzodiazepines, opiates, or cocaine; more than 7 days of inpatient treatment
`for substance abuse during the month prior to screening; or use of opiates, oral
`naltrexone, or disulfiram during the two weeks prior to screening.
`Detoxification prior to randomization was performed only if indicated by
`investigator judgment and had to be completed seven days before initiation of study
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 13 -
`
`therapy. Use of benzodiazepines was prohibited during the week prior to the first dose
`of study medication.
`Randomization procedures
`Patients were randomized to one of three treatment groups: long-acting
`injectable naltrexone 380 mg, long-acting injectable naltrexone 190 mg, or matching
`volumes of placebo (one-half of the placebo patients received an injection volume
`corresponding to 380 mg and the other halfreceived an injection volume
`corresponding to 190 mg). The study used a dynamic randomization procedure to
`balance allocation on gender, patient-specified goal of total abstinence, self-reported
`abstinence for the 7-day period prior to first injection, and study site.
`
`5
`
`10
`
`15
`
`Study Procedures and Outcome Definitions
`Patients received an injection of study medication at 4-week intervals over 24
`weeks, alternating between the left and right gluteus maximus. Injections were
`prepared and administered by individuals who were not involved in any of the safety
`or efficacy assessments, and treatment assignment was blinded to all study personnel.
`All patients received standardized supportive therapy (12 sessions) using the
`BRENDA model (Volpicelli JR, Pettinati HM, McLellan AT, O'Brien CP. Combining
`medication and psychosocial. treatments for addictions: the BRENDA approach. New
`20 York: The Guilford Press; 2001), a six-stage low-intensity intervention designed to
`facilitate direct feedback with respect to addiction-related consequences. During this
`trial, BRENDA sessions were administered by study site personnel including
`psychologists, nurses, therapists, counselors, and physicians.
`The number of standard drinks consumed per day was recorded using the
`Timeline Follow Back (TLFB) method (Sobell LC, Sobell MB. Timeline Followback:
`a technique for assessing self-reported ethanol consumption. In: Allen J, Litten RZ,
`eds. Measuring alcohol consumption: psychosocial and biological methods. Totowa,
`NJ: Humana Press, 1992:41-72). Breath alcohol levels of ~0.02 gm/dL were required
`before self-report data were collected. Patients who discontinued study drug treatment
`
`25
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 14 -
`
`prematurely were allowed to remain in the study, continue to follow the established
`visit and procedure schedule, and receive BRENDA treatment.
`
`Study Formulation: Naltrexone long-acting injection (Vivitrex®) consists of
`5 microspheres (approximately 100 µM) composed of naltrexone and polylactide-co-
`glycolide polymeric matrix (Medisorb®), a common biodegradable medical polymer
`with an extensive history of human use in absorbable sutures and extended-release
`pharmaceuticals. Following injection, naltrexone on the surface of the microspheres is
`released, yielding peak concentrations within three days. Thereafter, by a combination
`of diffusion and erosion, naltrexone is further released for greater than 30 days.
`
`10
`
`15
`
`Definition of Outcomes. The primary efficacy endpoint was the event rate of
`heavy drinking over the 24 weeks of treatment. The definition of heavy drinking (~5
`drinks per day for men and ~4 drinks per day for women) is consistent with that used
`in previous trials of oral naltrexone. Secondary endpoints included the event rate of
`"risky" drinking (>2 drinks per day for men and> 1 drink per day for women)
`specified by the National Institute on Alcohol Abuse and Alcoholism (NIAAA) (U.S.
`Department of Agriculture/U.S. Department of Health and Human Services. Home
`and Garden Bulletin no. 232. Nutrition and yo.ur health: dietary guidelines for
`20 Americans. 3rd ed. Washington, DC: Supt. of Docs., U.S. Government. Printing
`Office, 1990) and the event rate of any drinking days. Exploratory endpoints included
`serum gamma-glutamyl transferase (GGT) changes over time, and time to patient
`discontinuation. Adverse events were coded using the MedDRA dictionary of
`preferred terms.
`
`25
`
`30
`
`Statistical Methods
`The primary analysis for the primary and secondary endpoints was performed
`on an intention-to-treat population (i.e., all randomized patients who received at least
`one dose of study medication). The primary objective was to determine whether
`treatment with long-acting naltrexone (at either 190 mg or 380 mg) decreased the
`
`AMN1032
`IPR of Patent No. 7,919,499
`
`

`

`4000.3010 us
`
`- 15 -
`
`event rate of heavy drinking relative to treatment with placebo injection. Statistical
`methods to analyze multiple drinking episodes in alcoholism treatment clinical trials
`have been described by Wang et al., Short of complete abstinence: an analysis
`exploration of multiple drinking episodes in alcoholism treatment trials. Alcohol Clin
`Exp Res. 2002;26: 1803-9. The primary analysis for the primary endp.oint was
`performed using a stratified recurrent event Andersen-Gill like model with robust
`variance estimation (Lin DY, Wei LJ, Yang I, and Ying Z. Semiparametric regression
`for the mean and rate functions of recurrent events. J Royal Stat Soc (B) 2000;62:
`711-30; SAS/STAT user's guide. Version 8. Cary, NC: SAS Institute, 1999:2596).
`The 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