throbber
ALKERMES EXHIBIT 2013
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`Page 1 of 16
`
`

`

`Docket No. FDA-2007—D—03 69
`
`June 2, 2014
`Page 2
`
`from alcohol in an outpatient setting prior to initiation of treatment, and for the prevention of
`relapse to opioid dependence following opioid detoxification. Vivitrol should be administered as
`
`part of a comprehensive management program that includes psychosocial support. The product
`is designed to deliver therapeutic levels of naltrexone both quickly and on a sustained basis, to
`help patients avoid relapse during the early stages of their therapy and then to provide
`maintenance levels of naltrexone over a full, 30—day dosing cycle.
`
`To achieve the release profile on which FDA’s approval is based, Vivitrol relies on a
`precise formulation of polylactide-co-glycolide (PLG) microspheres, as well as a delivery system
`that deposits the microspheres locally at the IM gluteal injection site. From the local depot site,
`the release of the drug is characterized by an initial peak approximately two hours after injection,
`followed by a second peak approximately two to three days after injection. Plasma concentration
`levels then begin a slow decline at or about day 14. The release of the drug is dependent on,
`among other factors, the specific composition and formulation of the microspheres and the
`accompanying diluent, manufacturing and product quality, and the dynamic interaction between
`the microspheres and the conditions at the IM injection and depot site.
`
`Alkermes appreciates this opportunity to comment on the 2014 Draft Guidance. The
`Company recognizes that in this newly revised version of the guidance, the Agency has made the
`significant addition of recommending that generic sponsors conduct an in vitro drug release
`study. Alkermes supports the addition of this new study. However, further amendment of the
`
`2014 Draft Guidance is needed to assure that any proposed generic products match Vivitrol’s
`
`proven safety and efficacy profile.
`
`EXECUTIVE SUMMARY
`
`Vivitrol is a long-acting, modified release inj ectable drug product that delivers its active
`ingredient in three clinically significant phases:
`
`0 An “initial phase,” during which naltrexone is immediately absorbed from the
`
`surface of the formulation’s microspheres into the systemic circulation;
`
`o A “hydration phase,” during which physical erosion of the microspheres begins,
`triggering the release of subsurface naltrexone; and
`
`o A “sustained release phase,” during which the microspheres steadily erode and
`
`allow a constant amount of drug to be absorbed.
`
`The multiphasic release of naltrexone from the Vivitrol microspheres ensures that
`circulating naltrexone levels are quickly raised to a therapeutic level and then sustained over the
`course of the month-long dosing interval. Vivitrol therefore strikes a careful balance between
`
`Page 2 of 16
`
`

`

`Docket No. FDA-2007—D—0369
`
`June 2, 2014
`Page 3
`
`to ensure therapeutic maintenance over the full, 30—day dosing cycle. Vivitrol is indicated for
`treatment of alcohol dependence in patients who are able to abstain from alcohol in an outpatient
`setting prior to initiation of treatment. Vivitrol is also indicated for the prevention of relapse to
`opioid dependence, following opioid detoxification.
`
`To help ensure that generic versions of Vivitrol are able to match the multiphasic release
`profile of Vivitrol, Alkermes believes that the 2014 Draft Guidance should be further revised in
`
`two key respects.
`
`First, consistent with Alkermes’ 2011 comments, the in vivo study included in the 2014
`Draft Guidance should be amended to include statistical analyses of additional pharmacokinetic
`(PK) parameters that are necessary to ensure equivalence to a multiphasic, modified release,
`depot product such as Vivitrol. In support of this request, Alkermes has completed a series of in
`silico (computer-based) simulations comparing Vivitrol against potential comparator
`formulations. These simulations found that the traditional in viva bioequivalence metrics (i. e.,
`Cmax, AUCo.t, and AUCOM) are inadequate and could lead to non-equivalent generic versions of
`Vivitrol passing the Agency’s proposed BE standards.
`
`Thus, to the extent the 2014 Draft Guidance relies solely on conventional BE measures,
`the Guidance is insufficient to ensure equivalence. Because matching Vivitrol’s in vivo release
`profile is critical for generic products, at least one additional PK parameter is needed to ensure
`true bioequivalence. As proposed herein, the 2014 Draft Guidance should be revised to include
`the partial AUC metric AUC10_23. This additional metric is able to detect significant differences
`in the segment of the concentration-time profile that is most susceptible to potential therapeutic
`equivalence deficiencies.
`
`Second, also consistent with Alkermes’ 2011 comments, FDA should revise the 2014
`
`Draft Guidance to ensure that no significant difference in injection site reactions would exist
`between Vivitrol and generic products. In particular, the Company asks the Agency to include
`an in vivo comparative study designed to assess local injection site safety for any proposed
`generic. This injection site study could be conducted as its own stand-alone study, or
`
`incorporated as part of the required in vivo PK study.
`
`We appreciate FDA’s continued efforts to develop guidance and to request public
`comment on recommended studies for generic drugs. In light of Alkermes’ experience with
`Vivitrol and with sustained release drug formulations, we submit the following comments on the
`2014 Guidance for the Agency’s consideration.
`
`Page 3 of 16
`
`

`

`Docket No. FDA-2007-D—0369
`
`June 2, 2014
`Page 4
`
`BACKGROUND
`
`A.
`
`
`Vivitrol
`
`As explained in Alkermes’ 2011 comment document, the Agency first approved Vivitrol
`on April 13, 2006, under new drug application (NDA) 21-897. Vivitrol is currently approved for
`(a) the treatment of alcohol dependence in patients who are able to abstain from alcohol in an
`outpatient setting prior to initiation of treatment, and (b) the prevention of relapse to opioid
`dependence, following opioid detoxification. In both cases, treatment with Vivitrol should be
`
`part of a comprehensive management program that includes psychosocial support.
`
`The active ingredient in Vivitrol, naltrexone, is microencapsulated in a high molecular
`weight 75/25 PLG polymer at a concentration of 337 mg of naltrexone per gram of microspheres.
`Physicians are instructed to administer the product as an IM gluteal injection, using only the
`needles provided, alternating sides for subsequent injections.4 The recommended dose is one
`injection every four weeks or once a month.
`
`Vivitrol is a long-acting, modified release inj ectable drug product that delivers its active
`ingredient in the following three clinically significant phases: an initial phase, during which
`naltrexone is immediately absorbed from the surface of the formulation’s microspheres into the
`systemic circulation; a hydration phase, during which physical erosion of the microspheres
`begins, triggering the release of subsurface naltrexone; and a sustained release phase, during
`which the microspheres steadily erode and allow a constant amount of drug to be absorbed. The
`
`extent of naltrexone released at each of these three clinically important phases is a carefully
`
`controlled manufacturing and formulation-dependent variable.
`
`B.
`
`In Vivo Bioeguivalence For IM Depot Products
`
`With regard to the in vivo bioequivalence of systemically absorbed drugs, FDA generally
`
`recommends that applicants conduct single dose crossover or parallel group PK studies,
`measuring the concentrations of the generic and reference drugs in the blood or plasma.
`Applicants then calculate a variety of measurements of the resulting drug concentration versus
`
`time curve, including (1) the maximum concentration, Cmax; (2) the time to maximum
`
`concentration, TmaX; and (3) the area under the curve (AUC), both from dosing until the last
`
`measured time point, AUCo_t, and extrapolated to infinity, AUC0_00. Cmax and Tmax are believed to
`
`reflect the rate of the drug’s absorption into the systemic circulation, and the AUC measures are
`
`believed to reflect the extent of the drug’s absorption.
`
`4 Different length administration needles are provided to accommodate varying body physiques, given the
`
`Page 4 of 16
`
`

`

`Docket No. FDA-2007-D-O369
`
`June 2, 2014
`Page 5
`
`FDA has acknowledged that there are certain drugs for which the statistical comparison
`of Cmax and AUC as described above are insufficient to ensure that generic products will be as
`safe and effective as the RLD.5 This is particularly so for products that are approved and labeled
`for extended-release and whose release properties depend on complex formulations designed to
`release drug over multiple phases.6 For example, FDA has recommended partial AUC analysis
`for products such as Metadate CD, Concerta, Ritalin LA, Adderall XR, Focalin XR, and Ambien
`CR.7 In citizen petition responses regarding these products, FDA has stated it will require as a
`condition of ANDA approval that the generic product be BE to the RLD at the 90% confidence
`
`interval using appropriately tailored partial AUC metrics in addition to the conventional metrics
`Cm... AUCO. and AUco-mf.8
`
`Products such as Vivitrol — long acting depot formulations that are injected, and that
`reside subcutaneously over the course of the dosing interval, releasing drug in a multiphasic and
`sustained fashion — are a particularly complex extended—release dosage form, and present a new
`and challenging BE problem. In the past 12 months, FDA has released revised BE guidance
`documents for several of these products. These revised guidances reflect FDA’s ongoing process
`of addressing the BE challenges such products pose. Collectively, these BE guidances
`underscore the general principle that additional measures should be required for such products,
`but that there is no “one—size—fits—all” approach.
`
`For example, in February 2014, in addition to the revised guidance for Vivitrol, FDA
`issued revised draft BE guidances for Trelstar (triptorelin pamoate for inj ectable suspension),
`Lupron Depot (leuprolide acetate for depot suspension), and Sandostatin LAR Depot (octreotide
`
`5 See, e. g., FDA Citizen Petition Response, Daytrana, Docket No. FDA-2012—P-0932 (Jan. 23, 2013) at 3 (“FDA
`recognizes that, under certain circumstances, it may be appropriate to use a partial AUC parameter to ensure
`comparable therapeutic effects”).
`
`6 See generally Briefing Package for the April 13, 2010 Meeting of the Advisory Committee for Pharmaceutical
`Science and Clinical Pharmacology, available at:
`hm;://www.fda.gov/AdvisogCommittees/CommitteesMeetingMaterials/Drugs/AdvisoryCommitteeforPharmaceutic
`alScienceandClinicalPharmacology/ucmZO1700.htm.
`
`7 See Draft Guidance on Methylphenidate Hydrochloride (Metadate CD) (Sept. 2012); Draft Guidance on
`Methylphenidate Hydrochloride (Concerta) (Sept. 2012); Draft Guidance on Methylphenidate Hydrochloride
`(Ritalin LA) (Nov. 2011); Draft Guidance on Amphetamine Sulfate; Dextroamphetamine Saccharate;
`Dextroamphetamine Sulfate (Adderall) (Sept. 2012); Draft Guidance on Dexmethylphenidate Hydrochloride
`(Focalin XR) (Mar. 2012); Guidance on Zolpidem (Ambien CR) (Final) (Oct. 2011). Available at
`hm;://www.fda.gov/Drugs/GuidanceComplianceRegulatogInformation/Guidances/ucm075207.htrn.
`
`8 See FDA Citizen Petition Response, Metadate CD and Concerta, Docket Nos. FDA-2004-P-0151 and FDA-2004-
`P-0290 (July 19, 2012); FDA Citizen Petition Response, Adderall XR, Docket No. FDA-2005-P-0120 (June 22,
`2012); FDA Citizen Petition Response, Ambien CR, Docket No. FDA-2007~P-0182 (Oct. 13, 2010). FDA has also
`required the use of partial AUC metrics in cases where the RLD displays time-dependent localization of absorption
`within the gastrointestinal tract. See FDA Citizen Petition Response, Asacol, Asacol HD and Pentasa, Docket Nos.
`
`Page 5 of 16
`
`

`

`Docket No. FDA-2007-D-0369
`
`June 2, 2014
`Page 6
`
`acetate for inj ectable suspension).9 In each of these cases, FDA has recognized that conventional
`analysis of in vivo PK data is insufficient to assure BE.
`
`In particular, for Trelstar, Lupron Depot, and Sandostatin LAR Depot, FDA has
`specifically recognized the need for partial AUC analysis. The dosing and release features of
`these depot products, along with the key aspects of FDA’s revised BE recommendations, are as
`follows:
`
`0 Trelstar is available in three strengths: 3.75 mg, 11.25 mg, and 22.5 mg. The
`product is administered through a single intramuscular injection, with a dosing
`interval of 4 weeks, 12 weeks, and 24 weeks respectively. In the revised
`guidance, FDA recommended a single-dose, in vivo PK study at each strength,
`
`with BE based on AUC“, in addition to conventional analysis of AUCo_t, AUC0_w,
`
`and Cmax, where AUC“ is the area under the PK curve from day 7 to the last
`
`sampling time point of the dosing interval.
`
`0 Lupron Depot is available in strengths of 3.75 mg, 7.5 mg, 11.75 mg, 15 mg, 22.5
`mg, 30 mg, and 45 mg, administered through intramuscular injection, with dosing
`intervals ranging from 1 month to 6 months. Again, FDA recommended single—
`dose, in vivo PK studies, with BB based on AUC“, in addition to the conventional
`metrics.
`
`o
`
`Sandostatin LAR Depot is available in strengths of 10 mg, 20 mg, and 30 mg, and
`is administered intragluteally every 4 weeks (10 mg and 20 mg) or every 8 weeks
`(30 mg). Here, FDA has recommended a single-dose, in vivo PK study at 30 mg
`with BE based on AUC0.23 and AUC28_56, in addition to the conventional
`measurements.
`
`In each of these cases, FDA has recognized that the features of these depot products
`require additional analysis to ensure therapeutic equivalency. Specifically, partial AUC tailored
`to the PK of the product is an integral part of demonstrating BE across the length of the dosing
`interval.
`
`Similarly, FDA issued revised BE recommendations for Invega Sustenna (paliperidone
`palmitate extended—release inj ectable suspension) in December 2013, and Risperdal Consta
`
`9 Draft Guidance on Triptorelin Pamoate (Trelstar) (Feb. 2014); Draft Guidance on Leuprolide Acetate (Lupron
`Depot) (3.75 mg, 7.5 mg, 11.25 mg, 15 mg, 22.5 mg) (Feb. 2014); Draft Guidance on Leuprolide Acetate (Lupron
`Depot) (30 mg, 45 mg) (Feb. 2014); Draft Guidance on Octreotide Acetate (Sandostatin LAR) (Feb. 2014).
`
`Page 6 of 16
`
`

`

`Docket No. FDA-2007—D—0369
`
`June 2, 2014
`Page 7
`
`(risperidone long acting injection) in August 2013.10 For these two products, FDA has
`recommended multiple-dose, steady-state, in vivo PK studies, with analysis of steady-state PK
`parameters Cmin ss, Cmax ss, and AUCimerdose, as well as percent fluctuation at steady state (as
`represented by the equation [100*(Cmax ss — Cmin ss)/Caverage ss]). For Risperdal Consta, FDA
`also recognized the need for an in vitro BE study in order to evaluate the product’s release
`properties. The revised guidance for Risperdal Consta therefore reflects FDA’s determination
`that statistical analysis of key time points in an in vitro release study, combined with a steady-
`state in vivo study, together provide the necessary basis for demonstrating BE.
`
`These developments represent renewed focus and consideration on the part of the Agency
`regarding the complex issues associated with demonstrating BE for long-acting depot products.
`They signal an informed recognition by the Agency of the need to assure adequate plasma
`exposure across the length of the dosing interval.
`
`RECOMMENDED CHANGES TO DRAFT GUIDANCE
`
`A.
`
`The AUC Metric AUC10_28 Should Be Added To The BE Analysis
`
`Vivitrol is a long—acting depot drug whose safety and efficacy is driven by a distinct,
`multiphasic release and absorption profile designed to maintain patients on therapeutic levels of
`naltrexone across the entire one month dosing interval. As detailed in our comments on the 2009
`Draft Guidance, Vivitrol’s single-dose PK profile is comprised of three clinically important
`phases. It has an initial phase during which naltrexone is immediately absorbed from the surface
`of the formulation’s microspheres into systemic circulation, followed by a hydration phase
`during which physical erosion of the microspheres begins, triggering the release of subsurface
`naltrexone leading to Cmax. This, in turn, is followed by the main sustained release phase, during
`which the microsphere formulation erodes in situ to allow a constant amount of drug to be
`absorbed, maintaining therapeutic plasma concentrations until the next scheduled monthly
`administration.
`
`A single intramuscular injection of Vivitrol will reside at the site of deposition for a
`_
`period of weeks, and the extent of naltrexone release at each of the three clinically important
`phases is a carefully controlled manufacturing and formulation—dependent variable.11 The timing
`
`10 Draft Guidance on Paliperidone Palmitate (Invega Sustenna) (Feb. 2014); Draft Guidance on Risperidone
`(Risperdal Consta) (Feb. 2014). Available at
`hm;://www.fda.gov/Drugs/GuidanceComplianceRegulatoglnformation/Guidances/ucm075207.htm.
`
`” For example, the release of excessive drug by a generic during the initial burst phase could be the result of a
`manufacturing process leading to a greater amount of unencapsulated naltrexone at the surface of the microsphere,
`and different solvents or other residues from the manufacturing process could cause injection site reactions resulting
`in local physiological changes that affect drug absorption such as a sharp drop in local pH. Moreover, Vivitrol
`shares the property of many extended release products in that absorption from the site of injection is slower than
`
`Page 7 of 16
`
`

`

`Docket No. FDA-2007-D-0369
`
`June 2, 2014
`
`Page 8
`
`and initiation of these phases is critical to the overall safety and effectiveness of the product. A
`proposed generic version of Vivitrol that exhibits a different release rate would interrupt the
`product’s carefully designed dosing intervals and may deleteriously interfere with clinically
`significant plasma-drug concentrations.
`
`Accordingly, as recognized by the Agency in the 2014 Draft Guidance, a combination of
`
`in vitro and in vivo BE studies are required in order to adequately evaluate the therapeutic
`equivalence of test and reference products. FDA is therefore correct to require a demonstration
`of equivalence in an in vitro study, covering the different release phases of the product.
`However, we believe the rate and extent of adsorption of Vivitrol cannot be adequately measured
`using an in vitro dissolution methodology alone, because of the complex physiological
`interactions between the product and surrounding tissues at the site of deposition, and the
`possibility of manufacturer—dependent differences. Nor can it be accounted for with FDA’s
`
`traditional BE parameters of Cmax and AUC. The in vivo study must be able to independently
`establish the rate and extent of absorption. Thus, in addition to an appropriately robust in vitro
`study, the Agency should recommend the use of additional BE parameters, as discussed below,
`
`consistent with its recent recommendations for similar products.
`
`Specifically, Alkermes developed an in silico population PK model for Vivitrol, allowing
`
`the Company to computationally simulate its PK profile and evaluate the robustness of various
`PK metrics in the context of a BB analysis.12 The validated model was used to conduct clinical
`trial simulations of in vivo BE studies of Vivitrol 380 mg against alternative naltrexone
`
`formulations designed to be either BE or non-BE to Vivitrol 380 mg. These simulations
`
`produced PK profiles of the reference drug and the comparator drug, enabling standard BE
`
`assessment based on measurement of particular PK metrics. That is, we found the averages of
`
`particular PK metrics for the subjects in the comparator and reference arms of the simulated PK
`studies, calculated their ratio, and then determined if the 90% confidence interval fell within the
`BE limit of 80-125%.13
`
`of the plasma curve is much more dependent on formulation than is the curve for an immediate release product
`where actual rate of elimination plays a larger part. See generally Yafiez, J.A., et al., Flip-flop pharmacokinetics —
`delivering a reversal ofdisposition: challenges and opportunities during drug development, THER DELIV. (2011)
`2(5): 643-672 (attached at Tab 2).
`
`12 A population PK model is well-suited for an analysis of Vivitrol’s pharmacokinetics. Population PK models can
`identify correlations between patient characteristics and observed drug concentrations, which allow the models to
`account for and simulate inter-subject variability. Food and Drug Administration, Guidancefor Industry:
`Population Pharmacokinetics at 2 (Feb. 1999),
`ht_tp://www.fda.gov/downloads/ScienceResearch/SpecialTopics/WomensHealthResearch/UCM133184.pdf.
`Alkermes previously submitted the results of a separate population PK analysis (report #ALK21-011) in support of
`Vivitrol’s original marketing application. Vivitrol Clinical Pharmacology and Biopharmaceutics Review(s) at p.6-7
`
`of PDF (Mar. 31, 2005), ht_tp://www.accessdata.fda.gov/drugsatfda docs/nda/2006/0218975000 ClinPharmedf.
`
`‘3 Food and Drug Administration, Guidancefor Industry: Statistical Approaches to Establishing Bioequivalence at 2
`
`Page 8 of 16
`
`

`

`Docket No. FDA—2007-D-0369
`
`June 2, 2014
`Page 9
`
`By observing the frequency at which a particular PK metric was able to correctly confirm
`or reject a comparator drug as BE in our simulations, we were able to quantify the ability of that
`metric to serve as an indicator of whether a comparator drug is BE to Vivitrol. Using this
`framework, we investigated two issues:
`
`0 Whether a standard BE analysis of Vivitrol’s PK profile — based on measurement
`of Cmax, AUCO-inf, and AUCo_t(28 days) — consistently and accurately identified
`naltrexone formulations that are BE to Vivitrol, as well as rejected naltrexone
`formulations that are non—BE to Vivitrol; and
`
`0 Whether a partial AUC metric would be a more accurate predictor of
`bioequivalence than standard BE measures. Specifically, we investigated whether
`measurement of AUC0_3, AUC3_10, AUC10-28, or AUC14_28 could serve as a more
`
`accurate metric to recognize naltrexone formulations that are BE to Vivitrol, as
`
`well as to reject naltrexone formulations that are non-BE to Vivitrol.
`
`Detailed information on the development and validation of our Vivitrol population PK
`model may be found in the Appendix to these comments. Briefly, we developed our model using
`standard population PK methodology.” To obtain parameter values, we fit the model to PK data
`from ALK21-005, a single-center, double-blind, placebo-controlled Alkermes study that
`evaluated the single- and repeat—dose pharmacokinetics of Vivitrol in healthy subjects. To
`account for inter-subj ect variability, we included in the model a stochastic component based on
`an analysis of individual patient demographics and their associated PK profiles from ALK21-
`005. A visual depiction of our model’s structure and a table of its parameter values may be
`found in the Appendix.
`
`We evaluated each PK metric in two stages. First, to assess the power of a PK metric to
`identify formulations of naltrexone that are BE to Vivitrol, we ran clinical trial simulations of the
`
`reference product versus the reference product — that is, we ran clinical trial simulations of
`Vivitrol 380 mg against itself, and then assessed the ability of each PK metric to correctly
`identify the formulations as BE in spite of the inter-subj ect variability. As seen in Figure 1, we
`found that Cmax had the lowest power to identify Vivitrol 380 mg as BE with itself, which is
`likely due to the high inter-subj ect variability observed for Cmax in ALK21-005. The AUCO-inf
`
`and AUC04 parameters had the highest power to recognize Vivitrol 380 mg as BE with itself.
`The partial AUC metrics — AUC0_3, AUC3_10, AUC10-23, and AUC14_23 — had varying intermediate
`degrees of power between Cmax and AUCO-inf to recognize Vivitrol 380 mg as BE with itself, with
`AUC3_10 and AUC10.28 performing better than the other partial AUC metrics.
`
`M W. Byon et al., Establishing Best Practices and Guidance in Population Modeling: An Experience With an
`Internal Population Pharmacokinetic Analysis Guidance, 2 CPT: PHARMACOMETRICS & SYSTEMS PHARMACOLOGY
`
`Page 9 of 16
`
`

`

`Docket No. FDA-2007-D-0369
`
`June 2, 2014
`
`Page 10
`
`1.00—
`
`Ill
`
`m
`$0.75»
`,_
`E
`8 o 50
`< .
`2
`g.
`3
`g 025‘
`
`—
`
`E
`
`l
`25
`
`I
`|
`75
`50
`Number of Individuals Per Arm
`
`160
`
`M tr‘c
`8 '
`-e- CMAX
`a. AUCO_inf
`l- AUOD_28
`+ nuco_3
`@- Aucs_1o
`.
`“
`» AUCIO_28
`- AUCI4_28
`
`Figure 1: Probability of concluding bioequivalence of Vivitrol 380 mg (Reference) to
`Vivitrol 380 mg (Test) based on naltrexone pharmacokinetic parameters
`
`In the next stage of our analysis, we assessed the ability of each PK metric to reject two
`theoretical comparator formulations of naltrexone that were unlikely to be therapeutically
`equivalent to Vivitrol 380 mg. We framed the PK profile of these theoretical naltrexone
`formulations to emphasize potential differences between the 190 mg strength of Vivitrol, which
`was not efficacious in a clinical study of alcohol dependence, and the FDA approved 380 mg
`dose strength of Vivitrol. A significant number of patients receiving Alkermes’ investigational
`naltrexone 190 mg in ALK21-005 had naltrexone concentrations below 1 ng/mL during the
`sustained release phase. Notably, data from a clinical study wherein hydromorphone challenges
`were administered to test the ability of Vivitrol to block opioid agonism (ALK21-004)
`demonstrated that naltrexone concentrations above 1 ng/mL were sufficient to provide blockade.
`These data were utilized to substantiate investigation, and ultimately FDA approval, of Vivitrol
`for the treatment of opioid dependence. Thus, to simulate naltrexone formulations that were
`
`non-BE to Vivitrol, we designed the two following theoretical naltrexone formulations with
`diminished naltrexone concentrations —— i.e., less than 1 ng/mL — during Vivitrol’s sustained
`release phase:
`
`0 Naltrexone double release (NTX-DR) had a biphasic profile with a more
`pronounced initial peak at about 2 days, a secondary peak occurring at
`approximately 7 days, and a subsequent decline with no tertiary peak; and
`
`o Naltrexone single release (NTX-SR) had a single primary peak at about 2 days.
`
`Profile comparisons of NTX-DR 380 mg and NTX-SR 380 mg to Vivitrol 380 mg are
`shown in Figures 2 and 3, respectively, with the dashed line indicating a naltrexone
`
`Page 10 of 16
`
`

`

`Docket No. FDA—2007-D-0369
`
`June 2, 2014
`Page 11
`
`naltrexone formulations have prolonged periods with naltrexone concentrations less than 1
`ng/mL towards the tail ends of their PK profiles. Thus, like Alkermes’ naltrexone 190 mg, we
`would expect the simulated comparator naltrexone formulations to show a lack of efficacy and
`thus be non-BE to Vivitrol 380 mg. In this context, a strong PK metric would have a high
`probability of rejecting the comparator naltrexone formulations as non-BE to Vivitrol 380 mg.
`
`formulation
`‘I-Mviuol
`1,.
`
`E NTX—DR
`
` 0
`
`1O
`
`20
`TIME (days)
`
`1?
`
`i Z
`
`g
`n
`
`E
`
`0 8oO
`
`Figure 2: Population mean derived naltrexone concentration following administration of
`
`Vivitrol 380 mg (Reference) and NTX-DR (Comparator)
`
`formulation
`.,
`,I-IVuvinoI
`:
`-. "TX—SR
`
`
`
`15
`
`3 i:
`
`10
`a
`:9
`h
`E
`fl)
`
`8 5o
`o
`
`O
`
`O
`
`10
`
`20
`um: (days)
`
`30
`
`40
`
`Figure 3: Population mean derived naltrexone concentration following administration of
`
`Vivitrol 380 mg (Reference) and NTX—SR (Comparator)
`
`As seen in Figures 4 and 5, our simulations of Vivitrol versus NTX-DR and NTX-SR,
`
`respectively, found that the standard BE metrics performed poorly in distinguishing Vivitrol
`
`Page 11 of 16
`
`

`

`Docket No. FDA-2007-D-0369
`
`June 2, 2014
`Page 12
`
`greater than 15 individuals per arm. This raises the possibility that FDA’s recommended in vivo
`BE test, which relies on measurement of the standard BE metrics, is prone to type 1 error — i.e.,
`FDA’s test carries the risk that it will accept an alternative naltrexone formulation as BE to
`Vivitrol even if the alternative formulation exhibits fundamentally different release properties
`and therapeutic deficit (subtherapeutic PK) in the latter part of the dosing interval. On the other
`hand, our simulations of Vivitrol versus NTX-DR and NTX-SR found that partial AUC metrics
`performed strongly in distinguishing Vivitrol from the comparator naltrexone formulations.
`Three of the partial AUC metrics we tested — AUC3_10, AUC10_23, and AUC14_28 — rejected NTX-
`DR and NTX-SR as BE to Vivitrol across our simulations regardless of the number of
`individuals per arm.15
`
`1.00 —
`
`NI
`
`n a
`
`8 0'75_
`o
`g.
`.3
`gaso—
`2
`E
`€025"
`E
`
`0.00 —
`
`.
`Metric
`ae-CMAx
`*AUODJnf
`l-Auco_2a
`+Auco_3
`E- AU03_10

`1 mamas
`u-AUC‘I4_28
`
`25
`
`7'5
`5‘0
`Number of Individuals Per Arm
`
`100
`
`Figure 4: Probability of falsely concluding that NTX-DR 380 mg (Comparator) is
`
`bioequivalent to Vivitrol 380 mg (Reference) using traditional and proposed
`pharmacokinetic parameters
`
`15 As a general rule, increased clinical trial sample size results in an increased probability — due to reduction in
`statistical variability — that BE criteria will be met. But as seen in figures 4 and 5, as more individuals were added to
`each study arm in clinical trial simulations of Vivitrol, there was a greater likelihood that certain BE metrics would
`falsely accept a non-bioequivalent product as bioequivalent. Specifically, as the number of individuals per arm
`increased, we observed a corresponding increase in probability that PK metrics that were not sensitive to naltrexone
`concentration changes relative to the therapeutic threshold of 1 ng/mL (such as AUCOM) would result in a false
`acceptance of comparator formulations as BE to the reference formulation. However, for BB metrics that were
`sensitive to naltrexone concentration changes relative to the therapeutic threshold of 1 ng/mL (such as AUC10.28), the
`probability of falsely accepting the comparator formulations as BE did not increase regardless of the number of
`
`Page 12 of 16
`
`

`

`Docket No. FDA-2007-D-0369
`
`June 2, 20 14
`Page 13
`
`1.00 —
`
`III
`in
`a
`8 015 _
`2
`a
`fl
`E 0.50—
`'3.
`E
`3 0‘25 *
`
`32n
`
`.
`
`0.00 —
`
`.
`Metric
`li- CMAX
`‘AUCOJnf
`I-Auco_2a
`+ AU00_3
`a-AU03_10
`; AUC! 0.28
`AUC14_28
`
`Number of Individuals Per Arm
`
`Figure 5: Probability of falsely concluding that NTX-SR 380 mg (Comparator) is
`bioequivalent to Vivitrol 380 mg (Reference) using traditional and proposed
`pharmacokinetic parameters
`
`As noted previously, AUC3_10 and AUC10.23 performed better than the other partial AUC
`metrics in recognizing Vivitrol 380 mg as BE with itself (Figure 1). When combined with their
`superior ability to distinguish Vivitrol from the comparator naltrexone formulations, AUCHO and
`AUC10_23 emerge as compelling candidates for an additional metric to measure as a necessary
`supplement to FDA’S existing standards to ensure BE.
`
`In particular, analysis of AUC10_28 for BE provides a statistically robust test of the
`sustained release phase of Vivitrol’s PK profile. Furthermore, this segment of the PK profile
`encompasses the part of the curve that we believe may be most critical to assuring therapeutic
`equivalence of a generic sustained release inj ectable formulation of naltrexone.
`
`As discussed above, Vivitrol is intended to maintain plasma levels at therapeutic levels
`over a one month dosing interval. It achieves this through its multiphasic release profile and
`sustained release properties. Specifically, the formulation allows a single 380 mg dose of
`naltrexone to release an adequate amount of drug almost immediately and to maintain plasma
`concentrations at a clinically effective level for the entire dosing interval. For example, our
`studies of naltrexone blockade of the hydromorphone opioid response showed that a single dose
`of 300 mg of Vivitrol can maintain plasma concentrations at or above 1 ng/mL at day 28.16 In
`contrast, subjects receiving a 150 mg dose of the same formulation showed plasma levels below
`1 ng/mL at day 28 in all subj ects.17 These data help to illustrate that effective naltrexone
`
`'6 Vivitrol Clinical Pharmacology and Biopharmaceutics Review(s) at p.14-17 ofPDF (Mar. 31, 2005),
`ht_tp://www.accessdata.fda.gov/drugsatfda docs/nda/2006/0218975000 C1inPharmR.pdf.
`
`Page 13 of 16
`
`

`

`Docket No. FDA-2007—D-0369
`
`June 2, 2014
`
`Page 14
`
`blockade is a function of plasma concentration, and that a dose above 300 mg is required in order
`to maintain plasma levels above 1 ng/mL at the end of the dosing interval.
`
`Further, in clinical studies comparing 380 mg and 190 mg versus placebo, the 190 mg
`dose failed to show effectiveness.18 The PK profile differences between the 380 mg and
`investigational 190

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