throbber
Guidance for Industry
`Bioavailability and Bioequivalence
`Studies for Orally Administered
`Drug Products — General
`Considerations
`
`DRAFT GUIDANCE
`This guidance document is being distributed for comment purposes only.
`
`Comments and suggestions regarding this draft document should be submitted within 30 days of
`publication in the Federal Register of the notice announcing the availability of the draft
`guidance. Submit comments to Dockets Management Branch (HFA-305), Food and Drug
`Administration, 5630 Fishers Lane, rm. 1061, Rockville, MD 20852. All comments should be
`identified with the docket number listed in the notice of availability that publishes in the Federal
`Register.
`
`For questions regarding this draft document contact (CDER) Aida Sanchez 301-827-5847.
`
`U.S. Department of Health and Human Services
`Food and Drug Administration
`Center for Drug Evaluation and Research (CDER)
`
`July 2002
`BP
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Guidance for Industry
`Bioavailability and Bioequivalence
`Studies for Orally Administered
`Drug Products — General
`Considerations
`
`Additional copies are available from:
`Office of Training and Communication
`Division of Drug Information, HFD-240
`Center for Drug Evaluation and Research
`Food and Drug Administration
`5600 Fishers Lane
`Rockville, MD 20857
`(Tel) 301-827-4573
` http://www.fda.gov/cder/guidance/index.htm
`
`U.S. Department of Health and Human Services
`Food and Drug Administration
`Center for Drug Evaluation and Research (CDER)
`
`July 2002
`BP
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`TABLE OF CONTENTS
`
`INTRODUCTION................................................................................................................. 1
`I.
`II. BACKGROUND ................................................................................................................... 2
`A. General .................................................................................................................................2
`B. Bioavailability.......................................................................................................................3
`C. Bioequivalence ......................................................................................................................4
`III. METHODS TO DOCUMENT BA AND BE...................................................................... 6
`A.
`Pharmacokinetic Studies.......................................................................................................6
`B.
`Pharmacodynamic Studies..................................................................................................10
`C. Comparative Clinical Studies..............................................................................................10
`D.
`In Vitro Studies...................................................................................................................10
`IV. COMPARISON OF BA MEASURES IN BE STUDIES................................................. 11
`V. DOCUMENTATION OF BA AND BE............................................................................. 12
`A.
`Solutions.............................................................................................................................12
`B.
`Suspensions.........................................................................................................................13
`C.
`Immediate-Release Products: Capsules and Tablets ...........................................................13
`D. Modified-Release Products..................................................................................................14
`E. Miscellaneous Dosage Forms...............................................................................................17
`VI. SPECIAL TOPICS.............................................................................................................. 18
`A.
`Food-Effect Studies.............................................................................................................18
`B. Moieties to Be Measured.....................................................................................................18
`C. Long Half-Life Drugs..........................................................................................................20
`D.
`First Point Cmax.................................................................................................................20
`E. Orally Administered Drugs Intended for Local Action.......................................................20
`F. Narrow Therapeutic Range Drugs ......................................................................................21
`ATTACHMENT A: General Pharmacokinetic Study Design and Data Handling ............. 22
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`GUIDANCE FOR INDUSTRY1
`
`Bioavailability and Bioequivalence Studies for Orally Administered
`Drug Products — General Considerations
`
`This draft guidance represents the Food and Drug Administration's (FDA's) current thinking on this topic.
`It does not create or confer any rights for or on any person and does not operate to bind FDA or the
`public. An alternative approach may be used if such approach satisfies the requirements of the applicable
`statutes and regulations.
`
`I.
`
`INTRODUCTION
`
`This guidance is intended to provide recommendations to sponsors and/or applicants planning to
`include bioavailability (BA) and bioequivalence (BE) information for orally administered drug
`products in investigational new drug applications (INDs), new drug applications (NDAs),
`abbreviated new drug applications (ANDAs), and their supplements. This guidance is a revision
`of the October 2000 guidance. This revised guidance changes recommendations regarding
`(1) study design and dissolution methods development, (2) comparisons of BA measures, (3) the
`definition of proportionality, and (4) waivers for bioequivalence studies. The guidance also
`makes other revisions for clarification. The revisions should provide better guidance to sponsors
`conducting BA and BE studies for orally administered drug products. This guidance contains
`advice on how to meet the BA and BE requirements set forth in part 320 (21 CFR part 320) as
`they apply to dosage forms intended for oral administration. 2 The guidance is also generally
`applicable to non-orally administered drug products where reliance on systemic exposure
`measures is suitable to document BA and BE (e.g., transdermal delivery systems and certain
`rectal and nasal drug products). The guidance should be useful for applicants planning to
`conduct BA and BE studies during the IND period for an NDA, BE studies intended for
`submission in an ANDA, and BE studies conducted in the postapproval period for certain
`changes in both NDAs and ANDAs.3
`
`1 This guidance has been prepared by the Biopharmaceutics Coordinating Committee in the Center for Drug
`Evaluation and Research (CDER) at the Food and Drug Administration (FDA).
`
`2 These dosage forms include tablets, capsules, solutions, suspensions, conventional/immediate release, and
`modified (extended, delayed) release drug products.
`
`3 Other Agency guidances are available that consider specific scale-up and postapproval changes (SUPAC) for
`different types of drug products to help satisfy regulatory requirements in part 320 and § 314.70 (21 CFR 314.70).
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`1
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`II.
`
`BACKGROUND
`
`A.
`
`General
`
`Studies to measure BA and/or establish BE of a product are important elements in
`support of INDs, NDAs, ANDAs, and their supplements. As part of INDs and NDAs for
`orally administered drug products, BA studies focus on determining the process by which
`a drug is released from the oral dosage form and moves to the site of action. BA data
`provide an estimate of the fraction of the drug absorbed, as well as its subsequent
`distribution and elimination. BA can be generally documented by a systemic exposure
`profile obtained by measuring drug and/or metabolite concentration in the systemic
`circulation over time. The systemic exposure profile determined during clinical trials in
`the IND period can serve as a benchmark for subsequent BE studies.
`
`Studies to establish BE between two products are important for certain changes before
`approval for a pioneer product in NDA and ANDA submissions, and in the presence of
`certain postapproval changes in NDAs and ANDAs. In BE studies, an applicant
`compares the systemic exposure profile of a test drug product to that of a reference drug
`product. For two orally administered drug products to be bioequivalent, the active drug
`ingredient or active moiety in the test product should exhibit the same rate and extent of
`absorption as the reference drug product.
`
`Both BA and BE studies are required by regulations, depending on the type of application
`being submitted. Under § 314.94, BE information is required to ensure therapeutic
`equivalence between a pharmaceutically equivalent test drug product and a reference
`listed drug. Regulatory requirements for documentation of BA and BE are provided in
`part 320, which contains two subparts. Subpart A covers general provisions, while
`subpart B contains 18 sections delineating the following general BA/BE requirements:
`
` Requirements for submission of BA and BE data (§ 320.21)
` Criteria for waiver of an in vivo BA or BE study (§ 320.22)
` Basis for demonstrating in vivo BA or BE (§ 320.23)
` Types of evidence to establish BA or BE (§ 320.24)
` Guidelines for conduct of in vivo BA studies (§ 320.25)
` Guidelines on design of single-dose BA studies (§ 320.26)
` Guidelines on design of multiple-dose in vivo BA studies (§ 320.27)
` Correlations of BA with an acute pharmacological effect or clinical evidence
`(§ 320.28)
` Analytical methods for an in vivo BA study (§ 320.29)
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`2
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
` Inquiries regarding BA and BE requirements and review of protocols by FDA
`(§ 320.30)
` Applicability of requirements regarding an IND application (§ 320.31)
` Procedures for establishing and amending a BE requirement (§ 320.32)
` Criteria and evidence to assess actual or potential BE problems (§ 320.33)
` Requirements for batch testing and certification by FDA (§ 320.34)
` Requirements for in vitro batch testing of each batch (§ 320.35)
` Requirements for maintenance of records of BE testing (§ 320.36)
` Retention of BA samples (§ 320.38)
` Retention of BE samples (§ 320.63)
`
`B.
`
`Bioavailability
`
`Bioavailability is defined in § 320.1 as:
`
`the rate and extent to which the active ingredient or active moiety
`is absorbed from a drug product and becomes available at the site
`of action. For drug products that are not intended to be absorbed
`into the bloodstream, bioavailability may be assessed by
`measurements intended to reflect the rate and extent to which the
`active ingredient or active moiety becomes available at the site of
`action.
`
`This definition focuses on the processes by which the active ingredients or moieties are
`released from an oral dosage form and move to the site of action.
`
`From a pharmacokinetic perspective, BA data for a given formulation provide an
`estimate of the relative fraction of the orally administered dose that is absorbed into the
`systemic circulation when compared to the BA data for a solution, suspension, or
`intravenous dosage form (21 CFR 320.25(d)(2) and (3)). In addition, BA studies provide
`other useful pharmacokinetic information related to distribution, elimination, the effects
`of nutrients on absorption of the drug, dose proportionality, linearity in pharmacokinetics
`of the active moieties and, where appropriate, inactive moieties. BA data may also
`provide information indirectly about the properties of a drug substance before entry into
`the systemic circulation, such as permeability and the influence of presystemic enzymes
`and/or transporters (e.g., p-glycoprotein).
`
`BA for orally administered drug products can be documented by developing a systemic
`exposure profile obtained from measuring the concentration of active ingredients and/or
`active moieties and, when appropriate, its active metabolites over time in samples
`collected from the systemic circulation. Systemic exposure patterns reflect both release
`of the drug substance from the drug product and a series of possible presystemic/systemic
`actions on the drug substance after its release from the drug product. Additional
`comparative studies should be performed to understand the relative contribution of these
`processes to the systemic exposure pattern.
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`3
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`One regulatory objective is to assess, through appropriately designed BA studies, the
`performance of the formulations used in the clinical trials that provide evidence of safety
`and efficacy (21 CFR 320.25(d)(1)). The performance of the clinical trial dosage form
`may be optimized, in the context of demonstrating safety and efficacy, before marketing
`a drug product. The systemic exposure profiles of clinical trial material can be used as a
`benchmark for subsequent formulation changes and may be useful as a reference for
`future BE studies.
`
`Although BA studies have many pharmacokinetic objectives beyond formulation
`performance as described above, it should be noted that subsequent sections of this
`guidance focus on using relative BA (referred to as product quality BA) and, in
`particular, BE studies as a means to document product quality. In vivo performance, in
`terms of BA/BE, may be considered to be one aspect of product quality that provides a
`link to the performance of the drug product used in clinical trials, and to the database
`containing evidence of safety and efficacy.
`
`C.
`
`Bioequivalence
`
`Bioequivalence is defined in § 320.1 as:
`
`the absence of a significant difference in the rate and extent to which the
`active ingredient or active moiety in pharmaceutical equivalents or
`pharmaceutical alternatives becomes available at the site of drug action
`when administered at the same molar dose under similar conditions in an
`appropriately designed study.
`
`As noted in the statutory definitions, both BE and product quality BA focus on the release
`of a drug substance from a drug product and subsequent absorption into the systemic
`circulation. For this reason, similar approaches to measuring BA in an NDA should
`generally be followed in demonstrating BE for an NDA or an ANDA. Establishing
`product quality BA is a benchmarking effort with comparisons to an oral solution, oral
`suspension, or an intravenous formulation. In contrast, demonstrating BE is usually a
`more formal comparative test that uses specified criteria for comparisons and
`predetermined BE limits for such criteria.
`
`1.
`
`IND/NDAs
`
`BE documentation may be useful during the IND or NDA period to establish links
`between (1) early and late clinical trial formulations; (2) formulations used in
`clinical trial and stability studies, if different; (3) clinical trial formulations and to-
`be-marketed drug product; and (4) other comparisons, as appropriate. In each
`comparison, the new formulation or new method of manufacture is the test
`product and the prior formulation or method of manufacture is the reference
`product. The determination to redocument BE during the IND period is generally
`left to the judgment of the sponsor, who may wish to use the principles of relevant
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`4
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`guidances (in this guidance, see sections II.C.3, Postapproval Changes, and III.D,
`In Vitro Studies) to determine when changes in components, composition, and/or
`method of manufacture suggest further in vitro and/or in vivo studies should be
`performed.
`
`A test product may fail to meet BE limits because the test product has higher or
`lower measures of rate and extent of absorption compared to the reference product
`or because the performance of the test or reference product is more variable. In
`some cases, nondocumentation of BE may arise because of inadequate numbers of
`subjects in the study relative to the magnitude of intrasubject variability, and not
`because of either high or low relative BA of the test product. Adequate design
`and execution of a BE study will facilitate understanding of the causes of
`nondocumentation of BE.
`
`Where the test product generates plasma levels that are substantially above those
`of the reference product, the regulatory concern is not therapeutic failure, but the
`adequacy of the safety database from the test product. Where the test product has
`levels that are substantially below those of the reference product, the regulatory
`concern becomes therapeutic efficacy. When the variability of the test product
`rises, the regulatory concern relates to both safety and efficacy, because it may
`suggest that the test product does not perform as well as the reference product,
`and the test product may be too variable to be clinically useful.
`
`Proper mapping of individual dose-response or concentration-response curves is
`useful in situations where the drug product has plasma levels that are either higher
`or lower than the reference product and are outside usual BE limits. In the
`absence of individual data, population dose-response or concentration-response
`data acquired over a range of doses, including doses above the recommended
`therapeutic doses, may be sufficient to demonstrate that the increase in plasma
`levels would not be accompanied by additional risk. Similarly, population dose-
`or concentration-response relationships observed over a lower range of doses,
`including doses below the recommended therapeutic doses, may be able to
`demonstrate that reduced levels of the test product compared to the reference
`product are associated with adequate efficacy. In either event, the burden is on
`the sponsor to demonstrate the adequacy of the clinical trial dose-response or
`concentration-response data to provide evidence of therapeutic equivalence. In
`the absence of this evidence, a failure to document BE may suggest the product
`should be reformulated, the method of manufacture for the test product should be
`changed, and/or the BE study should be repeated.
`
`2.
`
`ANDAs
`
`BE studies are a critical component of ANDA submissions. The purpose of these
`studies is to demonstrate BE between a pharmaceutically equivalent generic drug
`product and the corresponding reference listed drug (21 CFR 314.94 (a)(7)).
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`5
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`Together with the determination of pharmaceutical equivalence, establishing BE
`allows a regulatory conclusion of therapeutic equivalence.
`
`3.
`
`Postapproval Changes
`
`Information on the types of in vitro dissolution and in vivo BE studies that should
`be conducted for immediate-release and modified-release drug products approved
`as either NDAs or ANDAs in the presence of specified postapproval changes is
`provided in the FDA guidances for industry entitled SUPAC-IR: Immediate
`Release Solid Oral Dosage Forms: Scale-Up and Post-Approval Changes:
`Chemistry, Manufacturing, and Controls, In Vitro Dissolution Testing, and In
`Vivo Bioequivalence Documentation (November 1995); and SUPAC-MR:
`Modified Release Solid Oral Dosage Forms: Scale-Up and Post-Approval
`Changes: Chemistry, Manufacturing, and Controls, In Vitro Dissolution Testing,
`and In Vivo Bioequivalence Documentation (September 1997). In the presence of
`certain major changes in components, composition, and/or method of manufacture
`after approval, in vivo BE should be redemonstrated. For approved NDAs, the
`drug product after the change should be compared to the drug product before the
`change. For approved ANDAs, the drug product after the change should be
`compared to the reference listed drug. Under section 506A(c)(2)(B) of the
`Federal Food, Drug, and Cosmetic Act (the Act) (21 U.S.C. 356a(c)(2)(B)),
`postapproval changes requiring completion of studies in accordance with part 320
`must be submitted in a supplement and approved by FDA before distributing a
`drug product made with the change.
`
`III. METHODS TO DOCUMENT BA AND BE
`
`As noted in § 320.24, several in vivo and in vitro methods can be used to measure product
`quality BA and establish BE. In descending order of preference, these include pharmacokinetic,
`pharmacodynamic, clinical, and in vitro studies. These general approaches are discussed in the
`following sections of this guidance. Product quality BA and BE frequently rely on
`pharmacokinetic measures such as AUC and Cmax that are reflective of systemic exposure.
`
`A.
`
`1.
`
`Pharmacokinetic Studies
`
`General Considerations
`
`The statutory definitions of BA and BE, expressed in terms of rate and extent of
`absorption of the active ingredient or moiety to the site of action, emphasize the
`use of pharmacokinetic measures in an accessible biological matrix such as blood,
`plasma, and/or serum to indicate release of the drug substance from the drug
`product into the systemic circulation. 4 This approach rests on an understanding
`
`4 If serial measurements of the drug or its metabolites in plasma, serum, or blood cannot be accomplished,
`measurement of urinary excretion may be used to document BE.
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`6
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`that measuring the active moiety or ingredient at the site of action is generally not
`possible and, furthermore, that some relationship exists between the
`efficacy/safety and concentration of active moiety and/or its important metabolite
`or metabolites in the systemic circulation. To measure product quality BA and
`establish BE, reliance on pharmacokinetic measurements may be viewed as a
`bioassay that assesses release of the drug substance from the drug product into the
`systemic circulation. A typical study is conducted as a crossover study. In this
`type of study, clearance, volume of distribution, and absorption, as determined by
`physiological variables (e.g. gastric emptying, motility, pH), are assumed to have
`less interoccasion variability compared to the variability arising from formulation
`performance. Therefore, differences between two products because of
`formulation factors can be determined.
`
`2.
`
`Pilot Study
`
`If the sponsor chooses, a pilot study in a small number of subjects can be carried
`out before proceeding with a full BE study. The study can be used to validate
`analytical methodology, assess variability, optimize sample collection time
`intervals, and provide other information. For example, for conventional
`immediate-release products, careful timing of initial samples may avoid a
`subsequent finding in a full-scale study that the first sample collection occurs after
`the plasma concentration peak. For modified-release products, a pilot study can
`help determine the sampling schedule to assess lag time and dose dumping. A
`pilot study that documents BE may be appropriate, provided its design and
`execution are suitable and a sufficient number of subjects (e.g., 12) have
`completed the study.
`
`3.
`
`Pivotal Bioequivalence Studies
`
`General recommendations for a standard BE study based on pharmacokinetic
`measurements are provided in Attachment A.
`
`4.
`
` Study Designs
`
`Nonreplicate study designs are recommended for BE studies of immediate-
`release and modified-release dosage forms. However, sponsors and/or applicants
`have the option of using replicate designs for BE studies for these drug products.
`Replicate study designs offer several scientific advantages compared to
`nonreplicate designs. The advantages of replicate study designs are that they (1)
`allow comparisons of within-subject variances for the test and reference products,
`(2) indicate whether a test product exhibits higher or lower within-subject
`variability in the bioavailability measures when compared to the reference
`product, (3) provide more information about the intrinsic factors underlying
`formulation performance, and (4) reduce the number of subjects needed in the BE
`study. The recommended method of analysis of nonreplicate or replicate studies
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`7
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`to establish BE is average bioequivalence, as discussed in section IV. General
`recommendations for nonreplicate study designs are provided in Attachment A.
`Recommendations for replicate study designs can be found in the Guidance for
`Industry Statistical Approaches to Establishing Bioequivalence (January 2001).
`
`5.
`
`Study Population
`
`Unless otherwise indicated by a specific guidance, subjects recruited for in vivo
`BE studies should be 18 years of age or older and capable of giving informed
`consent. This guidance recommends that in vivo BE studies be conducted in
`individuals representative of the general population, taking into account age, sex,
`and race. If the drug product is intended for use in both sexes, the sponsor should
`attempt to include similar proportions of males and females in the study. If the
`drug product is to be used predominantly in the elderly, the sponsor should
`attempt to include as many subjects of 60 years of age or older as possible. The
`total number of subjects in the study should provide adequate power for BE
`demonstration, but it is not expected that there will be sufficient power to draw
`conclusions for each subgroup. Statistical analysis of subgroups is not
`recommended. Restrictions on admission into the study should generally be
`based solely on safety considerations. In some instances, it may be useful to
`admit patients into BE studies for whom a drug product is intended. In this
`situation, sponsors and/or applicants should attempt to enter patients whose
`disease process is stable for the duration of the BE study. In accordance with
`§ 320.31, for some products that will be submitted in ANDAs, an IND may be
`required for BE studies to ensure patient safety.
`
`6.
`
`Single-Dose/Multiple-Dose Studies
`
`Instances where multiple-dose studies may be useful are defined under
`§ 320.27(a)(3). However, this guidance generally recommends single-dose
`pharmacokinetic studies for both immediate- and modified-release drug products
`to demonstrate BE because they are generally more sensitive in assessing release
`of the drug substance from the drug product into the systemic circulation (see
`section V). If a multiple-dose study design is important, appropriate dosage
`administration and sampling should be carried out to document attainment of
`steady state.
`
`7.
`
`Bioanalytical Methodology
`
`Bioanalytical methods for BA and BE studies should be accurate, precise,
`selective, sensitive, and reproducible. A separate FDA guidance entitled
`Bioanalytical Method Validation (May 2001) is available to assist sponsors in
`validating bioanalytical methods.
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`8
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`8.
`
`Pharmacokinetic Measures of Systemic Exposure
`
`Both direct (e.g., rate constant, rate profile) and indirect (e.g., Cmax, Tmax, mean
`absorption time, mean residence time, Cmax normalized to AUC)
`pharmacokinetic measures are limited in their ability to assess rate of absorption.
`This guidance, therefore, recommends a change in focus from these direct or
`indirect measures of absorption rate to measures of systemic exposure. Cmax and
`AUC can continue to be used as measures for product quality BA and BE, but
`more in terms of their capacity to assess exposure than their capacity to reflect
`rate and extent of absorption. Reliance on systemic exposure measures should
`reflect comparable rate and extent of absorption, which in turn should achieve the
`underlying statutory and regulatory objective of ensuring comparable therapeutic
`effects. Exposure measures are defined relative to early, peak, and total portions
`of the plasma, serum, or blood concentration-time profile, as follows:
`
`a. Early Exposure
`
`For orally administered immediate-release drug products, BE may generally be
`demonstrated by measurements of peak and total exposure. An early exposure
`measure may be informative on the basis of appropriate clinical efficacy/safety
`trials and/or pharmacokinetic/pharmacodynamic studies that call for better control
`of drug absorption into the systemic circulation (e.g., to ensure rapid onset of an
`analgesic effect or to avoid an excessive hypotensive action of an
`antihypertensive). In this setting, the guidance recommends use of partial AUC as
`an early exposure measure. The partial area should be truncated at the population
`median of Tmax values for the reference formulation. At least two quantifiable
`samples should be collected before the expected peak time to allow adequate
`estimation of the partial area.
`
`b.
`
`Peak Exposure
`
`Peak exposure should be assessed by measuring the peak drug concentration
`(Cmax) obtained directly from the data without interpolation.
`
`c.
`
`Total Exposure
`
`For single-dose studies, the measurement of total exposure should be:
`
`
`
`
`
`Area under the plasma/serum/blood concentration-time curve from time
`zero to time t (AUC0-t), where t is the last time point with measurable
`concentration for individual formulation.
`
`Area under the plasma/serum/blood concentration-time curve from time
`zero to time infinity (AUC0-), where AUC0- = AUC0-t + Ct/z, Ct is the
`last measurable drug concentration and z is the terminal or elimination
`
`J:\!GUIDANC\4964dft.doc
`07/10/02
`
`9
`
`AMN1069
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Draft — Not for Implementation
`
`rate constant calculated according to an appropriate method. The terminal
`half-life (t1/2) of the drug should also be reported.
`
`For steady-state studies, the measurement of total exposure should be the area
`under the plasma, serum, or blood concentration-time curve from time zero to
`time • over a dosing interval at steady state (AUC0-•), where • is the length of the
`dosing interval.
`
`B.
`
`Pharmacodynamic Studies
`
`Pharmacodynamic studies are not recommended for orally administered drug products
`when the drug is absorbed into the systemic circulation and a pharmacokinetic approach
`can be used to assess systemic exposure and establish BE. However, in those instances
`where a pharmacokinetic approach is not possible, suitably validated pharmacodynamic
`methods can be used to demonstrate BE.
`
`C.
`
`Comparative Clinical Studies
`
`Where there are no other means, well-controlled clinical trials in humans may be useful
`to provide supportive evidence of BA or BE. However, the use of comparative clinical
`trials as an approach to demonstrate BE is generally considered insensitive and should be
`avoided where possible (21 CFR 320.24). The use of BE studies with clinical trial
`endpoints may be appropriate to demonstrate BE for orally administered drug products
`when measurement of the active ingredients or active moieties in an accessible biological
`fluid (pharmacokinetic approach) or pharmacodynamic approach is infeasible.
`
`D.
`
`In Vitro Studies
`
`Under certain circumstances, product quality BA and BE can be documented using in
`vitro approaches (21 CFR 320.24(b)(5) and 21 CFR 320.22(d)(3)). For highly soluble,
`highly permeable, rapidly dissolving, orally administered drug products, documentation
`of BE using an in vitro approach (dissolution studies) is appropriate based on the
`biopharmaceutics classification system. 5 This approach may also be suitable under some
`circumstances in assessing BE during the IND period, for NDA and ANDA submissions,
`and in the presence of certain postapproval changes to approved NDAs and ANDAs. In
`addition, in vitro approaches to document BE for nonbioproblem drugs approved before
`1962 remain acceptable (21 CFR 320.33).
`
`Dissolution testing is also used to assess batch-to-batch quality, where the dissolution
`tests, with defined procedures and acceptance criteria, are used to allow batch release.
`Di

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