throbber
DEVELOPING
`
`SOLID ORAL
`
`DOSAGE FORMS:
`
`PHARMACEUTICAL
`
`THEORY AND
`
`PRACTICE
`
`Executive editors
`
`Yihong Qiu, Abbott Laboratories, IL, USA
`
`Yisheng Chen, Novast Laboratories, Nantong, China
`
`Geoff G. Z. Zhang, Abbott Laboratories, IL, USA
`
`Associate editors
`
`Lirong Liu, Pfizer Inc, N], USA
`
`William R. Porter, Abbott Laboratories, IL, USA
`
` AMSTERDAM 6 BOSTON 0 HEIDELBERG 0 LONDON 0 NEW YORK 0 OXFORD
`
`PARIS 0 SAN DIEGO 0 SAN FRANCISCO 0 SINGAPORE o SYDNEY 0 TOKYO
`
`Academic Press is an imprint of Elsevier
`
`InnoPharma Exhibit 1023.0001
`
`

`

`Academic Press is an imprint of Elsevier
`30 Corporate Drive, Suite 400, Burlington, MA 01803, USA
`32 Jamestown Road, London NW1 7BY, UK
`525 B Street, Suite 1900, San Diego, CA 92101—4495, USA
`360 Park Avenue South, New York, NY 10010-1710, USA
`
`First edition 2009
`
`Copyright © 2009 Elsevier Inc. Apart from Chapters 12 and 38 which are in the public domain. All rights reserved.
`
`No part of this publication may be reproduced, stored in a retrieval system or transmitted in any form or by any means
`electronic, mechanical, photocopying, recording or otherwise without the prior written permission of the publisher
`
`Permissions may be sought directly from Elsevier’s Science & Technology Rights Department in Oxford, UK: phone
`(+44) (0) 1865 843830; fax (+44) (0) 1865 853333; email: permissions@elsevier.com. Alternatively you can submit your
`request online by visiting the Elsevier web site at http: / / elsevier.com / locate / permissions, and selecting Obtaining
`permission to use Elsevier material
`
`Notice
`
`No responsibility is assumed by the publisher for any injury and / or damage to persons or property as a matter of prod—
`ucts liability, negligence or otherwise, or from any use or operation of any methods, products, instructions or ideas con-
`tained in the material herein.
`
`
`
`Disclaimer
`
`
`
`
`Medicine is an ever-changing field. Standard safety precautions must be followed, but as new research and clinical
`experience broaden our knowledge, changes in treatment and drug therapy may become necessary or appropriate.
`Readers are advised to check the most current product information provided by the manufacturer of each drug to be
`administered to verify the recommended dose, the method and duration of administrations, and contraindications. It
`is the responsibility of the treating physician, relying on experience and knowledge of the patient, to determine dos-
`ages and the best treatment for each individual patient. Neither the publisher nor the authors assume any liability for
`any injury and / or damage to persons or property arising from this publication.
`
`Library of Congress Cataloging-in-Publication Data
`A catalog record for this book is available from the Library of Congress
`
`British Library Cataloguing in Publication Data
`A catalogue record for this book is available from the British Library
`
`ISBN: 978-0-444-53242-8
`
`
`For information on all Academic Press publications
`Visit our website at www.Elsevierdirect.com
`
`Typeset by Charon Tec Ltd., A Macmillan Company
`www.macmillansolutions.com
`
`Printed and bound in the United States of America
`
`0910111213 10987654321
`
`
`Working together to grow
`libraries in developing countries
`www.clsevier.com I WWW.b00kaid.0rg | www.52bre.org
`Sabre Foundation
`ELSEVIER
`£295,353
`
`InnoPharma Exhibit 1023.0002
`
`

`

`
`
`This material may be protected by Copyright law (Title 17 U5. Code)
`
`
`
`CHAPTER
`
`15
`
`Bioavailability and Bioequivalence
`
`Hao Zhu, Honghui Zhou and Kathleen Seitz
`
`15.1 GENERAL BACKGROUND
`
`Substituting one oral dosage formulation for
`another has been a common practice for many years
`in the drug development industry, as well as in the
`clinic. During drug development, for example, indus-
`try scientists frequently evaluate different versions of
`investigational drug products or dosage formulations,
`and thereby often need to conduct bioequivalence
`studies. In turn, prescribing clinicians use the bio-
`availability and bioequivalence data provided on the
`product label to select an optimal treatment regimen
`for their patients. Consequently, the overall therapeu-
`tic success of any drug substitution in the clinic will
`ultimately depend on multiple factors. These factors
`include the pharmacokinetics of the comparator and
`reference drugs, as well as the appropriateness of the
`study design and statistical criteria that were used to
`initially demonstrate bioequivalence.
`Bioequivalence has evolved as a specific regulatory
`requirement over the last 40 years. In the early 19605,
`researchers noticed that the bioavailability of a thera-
`peutic drug product could vary depending on its oral
`dosage formulation.1 Almost 15 years later, in 1977,
`the US Food and Drug Administration (FDA) initially
`published their recommended procedures for spon-
`sors to use in studies of bioequivalence.1 Since then,
`the FDA has continued to refine and revise its guid-
`ance on bioequivalence, other international regula-
`tory agencies have published their own guidelines,
`and pharmaceutical
`industry professionals world-
`wide have contributed their statistical expertise and
`
`scientific opinions. Nevertheless, several major con-
`troversies have yet to be resolved. Issues pertaining to
`the selection of the most appropriate statistical criteria
`to use to sufficiently demonstrate bioequivalence are
`still frequently debated. Other topics of discussion
`include a determination of exactly when (or under
`what specific circumstances) should formal tests of
`bioequivalence be required from a regulatory stand-
`point, and for which type or class of drug.
`Under the Food, Drug and Cosmetic (FD&C) Act of
`1938 and the 1962 Kefauver—Harris Amendment, spon-
`sors were required to provide safety and efficacy data
`to support all claims for the active ingredients in a
`new drug product before it could be approved for sale.
`Scientific standards later set by the FDA, however,
`have since allowed sponsors to lawfully make appro-
`priate drug substitutions without necessarily having
`to conduct additional
`time-consuming, and expen-
`sive, clinical safety and efficacy studies. That is, in the
`absence of additional clinical studies (and under spe-
`cific circumstances), an appropriate set of biopharma-
`ceutical, pharmacokinetic, and statistical evaluations
`may now be used to establish that a test drug formu-
`lation is bioequivalent (and thereby therapeutically
`interchangeable) with a reference drug formulation.
`The information included in this chapter will pro—
`vide the reader with an overview of the clinical, phar-
`macokinetic, and statistical
`issues associated with
`
`bioavailability and bioequivalence studies of oral
`dosage formulations. The current international regu-
`latory perspectives will also be presented, along with
`a detailed comparison of the various criteria presently
`
`Dwelnpmg Solid Oral Dosage Forms Pharmaceutical Theory and Practice
`lSBN: 978»0—444—53242.8
`
`341
`
`Copyright © 200‘). Elscvwr Inc.
`All rights reserved
`
`InnoPharma Exhibit 1023.0003
`
`

`

`
`
`342
`
`15, BIOAVAILABILITY AND BIOEQUIVALENCE
`
`being used in the pharmaceutical industry to demon-
`strate bioequivalence.
`
`15.2 DEFINITIONS AND KEY CONCEPTS
`
`Bioavailability essentially describes the overall rate
`and extent of drug absorption. Data from bioavail-
`ability studies are routinely used to identify a drug
`product’s pharmacokinetics, optimize a therapeutic
`dose regimen, and support product labeling require-
`ments. Bioequivalence, on the other hand, gener—
`ally describes the extent to which the bioavailability
`of one particular drug product (i.e., the test product)
`compares with that of another known drug product
`(i.e., the reference product). Data from bioequivalence
`studies are often used to establish a link between dif-
`
`ferent investigational drug formulations (e.g., an early
`phase 1 formulation versus a later phase 3 formula-
`tion). Bioequivalence studies may also be required
`during the post-approval period in certain situations,
`such as whenever a major change occurs in a manu-
`facturing method for an approved drug product.
`Bioequivalence studies are also generally required to
`compare generic versions of a drug product with the
`corresponding reference-listed drug.
`Regulatory requirements
`for bioavailability and
`bioequivalence data submitted with new drug applica-
`tions (NDAs) and supplemental applications are specifi-
`cally addressed in the US Code of Federal Regulations,2
`and a corresponding FDA guidance document has
`been published.3 The following sections will provide an
`overview of the key concepts, and general underlying
`principles, of bioavailability and bioequivalence.
`
`15 .2.1 Bioavailability
`
`When a drug is administered orally (or by any
`other extravascular route), a sufficient amount of the
`administered dose must be absorbed over a certain
`
`time period before the intended pharmacologic effect
`can manifest. Thus, the bioavailability of an orally
`administered drug clearly depends on a combination
`of factors,
`including the physiochemical character-
`istics of the drug formulation, and the physiological
`state of the gastrointestinal (GI) system.
`Bioavailability of an oral dosage form is defined3 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"
`
`two specific
`From a pharmacokinetic perspective,
`types of bioavailability can be considered: absolute
`
`bioavailability, and relative bioavailability. Absolute
`bioavailability is a special case in which the systemic
`exposure of an extravascular dosage form is deter-
`mined relative to that of its intravenous (IV) dosage
`form. Relative bioavailability, in contrast, compares
`the rate and extent of absorption of one dosage for-
`mulation (e.g., oral solution) to another dosage formu-
`lation (e.g., oral capsule). Relative bioavailability can
`also sometimes compare the rate and extent of absorp-
`tion for one drug product with two different adminis-
`tration routes (e.g., intramuscular and subcutaneous).
`For the most part, absolute bioavailability is gen—
`erally determined by comparing the extent of drug
`absorption after an extravascular versus an intrave-
`nous (e.g.,
`infusion or bolus) administration. Thus,
`valid extravascular and IV data are both typically
`required for the calculation of absolute bioavailability.
`In operational terms, this means two series of phar-
`macokinetic samples must be collected—one extravas-
`cular series, and one IV series—using a suitable
`biological matrix (e.g., blood, plasma or serum), and
`appropriate sampling schedules. In addition, drug
`concentrations from each series must be analyzed
`using a validated drug assay.
`Measured concentration data from each series are
`
`then plotted, and the area under the drug concentration—
`time curves (AUC) estimated (e.g., by applying a
`numerical integration formula such as the trapezoidal
`rule). Assuming clearance remains constant AUC
`is directly proportional
`to the amount of drug
`absorbed. Thus absolute oral bioavailability (P) can be
`calculated:
`
`
`2 Div Aucpo
`DP,
`‘ ALICw
`
`where:
`
`Div and DPO are the intravenous and oral doses
`administered, respectively
`AUC,,, and AUC},0 are the AUC estimates for the
`intravenous and oral routes, respectively.
`
`In contrast to absolute bioavailability, relative bio-
`availability essentially compares the rate and extent of
`absorption of one dosage formulation (e.g., oral solu-
`tion) to that of another (e.g., oral capsule). Over the
`course of a typical drug development cycle, several
`relative bioavailability studies could potentially be
`required (e.g., to compare the in viva performance of an
`earlier stage formulation versus the later stage formu-
`lation). Depending on the overall pace of drug devel-
`opment, new dosage formulations are often still being
`prepared while a new molecular entity progresses
`from the nonclinical stage into the early clinical stage.
`In cases where the final product is intended as a solid
`
`I], BIOPHARMACEUTICAL AND PHARMACOKINETIC EVALUATIONS OF DRUGS MOLECULES AND DOSAGE FORMS
`
`InnoPharma Exhibit 1023.0004
`
`

`

`
`
`15.2 DEFINITIONS AND KEY CONCEPTS
`
`343
`
`early drug exposure. Cmax is used to describe peak
`exposure. For a single dose study, both AUC0_T and
`AUCL-Hc are used to measure the total exposure. If a
`multiple dose study is appropriate, the AUCO,T at
`steady state (where T is the dosing interval) is used to
`describe total exposure.
`Pharmacodynamic assessments may also some-
`times be performed. For instance, if the systemic expo-
`sure of the drug is too low to be reliably detected or if
`an appropriate bioanalytical methodology cannot be
`developed to support a pharmacokinetic assessment,
`an appropriate pharmacodynamic assessment (i.e., a
`reliable and predictable surrogate marker) may suf-
`fice. Pharmacodynamic measurements are usually not
`recommended if pharmacokinetic measurements are
`available.
`
`Pharmacokinetic bioequivalence methods can be
`quite challenging for drugs with minimal systemic bio-
`availability. Drug classes that typically show minimal
`systemic bioavailability are ophthalmic, dermal, intra—
`nasal, and inhalation drugs. Nevertheless, pharmaco-
`dynamic assessments are not routinely used to show
`bioequivalence, for several reasons. First, very few val-
`idated, predictable surrogate biomarkers are available
`that are also considered acceptable surrogates by the
`regulatory authorities. Secondly, pharmacodynamic
`studies generally require prohibitively large sample
`sizes since intra- and inter-subject variability levels
`tend to be relatively high. Some examples of biological
`markers that have been successfully used for bioequiv-
`alence testing are skin blanching4 with corticosteroids,
`and stomach acid neutralization with antacids.5
`
`15.2.3 Pharmaceutical Equivalence and
`Therapeutic Equivalence
`
`Drug products are considered to be pharmaceuti-
`cal equivalents if they contain the same active ingredi-
`ent, in the same amount, with identical dosage forms,
`and identical routes of administration. Furthermore,
`
`drug products are considered to be therapeutically
`equivalent only if they are pharmaceutical equivalents
`(as described above) that are expected to produce the
`same clinical effects, and have similar safety profiles
`when they are administered to patients under the
`same conditions as specified in the product labeling
`information.
`
`Therapeutic equivalence is thus an ultimate meas-
`ure of the interchangeability of two distinct drug
`products or formulations. Therapeutic equivalence
`may be reasonably inferred from results of appropri-
`ately designed in viva pharmacokinetic bioequivalence
`studies.
`
`oral dosage form, for example, oral solutions or sus-
`pensions might be the only formulations ready for
`use. Solid prototype formulations, such as capsules,
`might also be ready for use in early phase 1; however
`these prototypes are often far from the final market-
`able form. Under these types of circumstances, there-
`fore, an estimate of the drug’s relative bioavailability
`is needed.
`
`Relative bioavailability (PM) can be calculated:
`
`A
`
`~ DA AUCB
`PM _ “[7 MC
`B
`
`where:
`
`DA and DB are the doses administered for drug for-
`mulation A and B, respectively
`AUCA and AUCB are the AUC estimates for the A
`and B formulations, respectively.
`
`15.2.2 Bioequivalence
`
`Bioequivalence is defined3 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 adminis—
`tered at the same molar dose under similar conditions in an
`
`appropriately designed study.”
`
`From a regulatory perspective, and for various stra-
`tegic reasons, bioequivalence studies may need to be
`conducted before a product is approved for use in the
`clinic or during the post—approval period. Depending
`on the particular research objective, for example, a
`pre-approval bioequivalence study might be required
`in an NDA submission to demonstrate the therapeutic
`link between the early phase dosage formulation and
`the to-be-marketed formulation. Bioequivalence stud-
`ies are also typically required in abbreviated NDAs
`for generic versions of brand name drugs.
`Two oral drug products can generally be considered
`to be bioequivalent if their respective concentration—
`time profiles are so similar that it would be unlikely
`that
`they would produce clinically significant dif-
`ferences in the pharmacological response. In other
`words, bioequivalent drug products with essentially
`the same systemic bioavailability should thus be
`able to produce similar, and predictable, therapeutic
`effects. Pharmacokinetic assessments in bioequiva-
`lence studies of solid oral drug products, therefore,
`typically include statistical comparisons of AUC and
`maximum concentration (Cmax). Different measures
`are sometimes needed to describe drug exposure.
`For example, partial AUC truncated to the median
`Tmax of the reference product can be used to describe
`
`11 BIOPHARMACEUTICAL AND PHARMACOKINETIC EVALUATIONS OF DRUGS MOLECULES AND DOSAGE FORMS
`
`InnoPharma Exhibit 1023.0005
`
`

`

`
`
`344
`
`15, BIOAVAILABILITY AND BIOEQUIVALENCE
`
`15.3 STATISTICAL CONCEPTS IN
`
`BIOEQU IVALENCE STUDIES
`
`A test (T) drug product and a reference (R) drug
`product can only be considered bioequivalent
`if
`equivalence of the rate and extent of drug absorption
`are demonstrated. This leads to three key questions:
`
`1. What are the pharmacokinetic measures that can
`be used to best characterize the rate and extent of
`
`drug absorption?
`2. What are the most appropriate statistical criteria to
`use to adequately demonstrate bioequivalence?
`3. How should a study be designed to sufficiently,
`and appropriately, capture the information
`necessary to show bioequivalence?
`
`Each of these major topics will be thoroughly dis-
`cussed in the next several sections.
`
`15.3.1 Selection and Transformation of
`
`Pharmacokinetic Measures
`
`In general, the pharmacokinetic measures that are
`typically used to describe the rate and extent of drug
`absorption include Cmax, time to reach Cmax (Tmax),
`and AUC. Whether this particular set of measures
`best reflects the actual rate and extent of drug absorp-
`tion, however, continues to be debated and discussed
`
`among many members of the industry and the regula-
`tory agencies.”
`The two most commonly used pharmacokinetic
`variables in bioequivalence studies are AUC, and
`Cmax. While Cmax, and Tmax, are both commonly used
`in standard in viva pharmacokinetic studies to reflect
`absorption rate, Cmax is specifically used in bioequiv-
`alence studies, whereas Tmax is not used as often
`because of the high interindividual variability typi-
`cally seen with this parameter. In addition to Cmax,
`AUC is also routinely used in bioequivalence studies.
`Assuming the in vivo clearance of a drug is constant
`for a given individual, AUC is directly proportional to
`the amount of drug being absorbed, and thus ideally
`describes the extent of drug absorption.
`To demonstrate equivalence,
`the mean AUC (or
`Cmax) values for two different products are directly
`compared. Mathematically, the easiest way is to do
`this is to calculate the difference between the two
`mean values. As an extension, the difference between
`
`the two log-transformed means can likewise be calcu-
`lated. Logarithmic transformation of AUC (or Cmax)
`provides several important advantages. A ratio com-
`paring log-transformed means can easily be converted
`
`back to a ratio of means in the normal scale, which can
`then be readily communicated to regulatory reviewers
`and prescribing clinicians. From a general clinical per-
`spective the ratio of the mean AUC (or Cmax), for the
`test versus the reference drugs, provides the most clin-
`ically relevant information. This is generally accepted
`in the industry, and has been recommended by the
`FDA Generic Drug Advisory Committee in 1991 .8'9’10
`From a pharmacokinetic perspective, pharma-
`cokinetic measures in bioequivalence studies can be
`expressed in a multiplicative fashion. AUC, for exam-
`ple, can be expressed as:
`
`F'D
`
`AUC0_OO :
`
`where:
`
`D is the dose given to a subject
`CL represents the clearance
`F is the bioavailability which is the fraction of the
`dose being absorbed (O S F S 1).
`
`This equation clearly shows that each subject’s AUC
`value is inversely proportional to a specific CL value.
`This multiplicative term (CL) can thus be regarded as a
`function of subject. However, in the statistical analysis of
`bioequivalence, all factors (e.g., treatment, subjects, and
`sequence) that contribute to the variation in pharmacoki-
`netic measurements are considered to be additive effects.
`
`Westlake11 contends that the subject effect is not additive,
`if data are analyzed on the original scale. Logarithmic
`transformation of AUC yields an additive equation:
`
`log AUCO,GO = log D + log P — log CL
`
`which is therefore preferable in bioequivalence analy-
`ses. Similar arguments can also be applied to Cmax.
`Finally, from a statistical perspective, AUC (or Cmax)
`values in bioequivalence studies are often positively
`skewed, and the variances between the test and refer-
`
`ence groups may differ. This generates an inconsist-
`ency in the two major assumptions (i.e., normality,
`and homoscedasticity of the variance) of the statisti-
`cal analysis of bioequivalence. Log-transformation
`of AUC (or Cmax) values, however, makes the distri-
`bution appear more symmetric, closer to the normal
`distribution, and achieves a relatively homogeneous
`variance. Thus, major international regulatory agen-
`cies generally recommend using log-transformations
`of pharmacokinetic measurements like AUC or Cmax in
`bioequivalence analyses. However, one main drawback
`to using log-transformation is that, although the mean
`log AUC (or log Cmax) values are compared between
`the test and reference groups,
`log-transformation
`essentially compares the median AUC (or Cmax) values
`instead of the means from each group.12
`
`II. BIOPHARMACEUTICAL AND PHARMACOKINETIC EVALUATIONS OF DRUGS MOLECULES AND DOSAGE FORMS
`
`InnoPharma Exhibit 1023.0006
`
`

`

`
`
`15.3 STATISTICAL CONCEPTS IN BIOEQUIVALENCE STUDIES
`
`15.3.2 Variability in Pharmacokinetic
`Measures of Bioavailability
`
`15.3.3 Statistical Criteria for Evaluating
`Bioequivalence
`
`The variability in pharmacokinetic measurements
`(e.g., log AUC or log Cmax values) of bioavailability
`can be represented in two hierarchical layers.13'14 That
`is, bioavailability measurements can vary at the indi-
`vidual subject level, and at overall population level.
`Whenever oral bioavailability measurements
`are
`repeated over time, for example, an individual sub-
`ject’s values may vary from time to time, even if the
`same drug product is administered each time. This
`series of measures can then collectively form a distri—
`bution where the mean value represents the inherent
`bioavailability, and the variance reflects the within—
`subject variability. Of note, in addition to the changes
`in subject-level information over time, any intra— or
`inter-lot variability arising from the manufacture of
`the drug product over time will also be reflected in
`the within—subject variability.
`The second hierarchical layer of variability rep-
`resents the overall population level variance. In the
`overall study population,
`individual subjects may
`have variations in their own inherent bioavailability.
`In all, these variations collectively form a distribu-
`tion where the mean represents the population aver-
`age of the bioavailability, and the variance reflects the
`between-subject variability. For example, in a non-
`replicated parallel bioequivalence study, patients are
`divided into two groups and given either a test (T) or
`reference I product. The jth subject in the trial would
`have a bioavailability measurement (e.g., log AUC or
`log Cmax) of Yij, where i represents the product being
`given (1' = T or R). Then Yq- follows a distribution with
`mean pi], and variance 0,2“ representing the within-
`subject variability for a given product. The individ-
`ual means m]- (j = T or R) are jointly distributed with
`mean M}, and variance of”
`(where 0%,
`is the between
`subject variability).
`The total variance for each formulation is then
`
`derived as the sum of the Within- and between-subject
`variability 0Tj = 0%}. + 0%,]:
`.
`This model allows the correlation (p), between MT
`and MB. Then the subject-by-formulation interaction
`variance component is defined as:
`
`e=meW—W)
`: (GET _ 03102 + 2'(1_ :0)'03T ‘OBR
`
`In general, a linear mixed-effect model is used to
`evaluate bioequivalence. For crossover designs or
`repeated measurements, the means are given additional
`structure by including period and sequence effects.
`
`The statistical tools (i.e., criteria) available for use
`in bioequivalence analyses have evolved remarkably
`over the past few decades. This has mostly occurred
`through a joint effort by interested stakeholders (i.e.,
`professionals
`in academia,
`industry, and regula-
`tory agencies) to select the most appropriate tools
`to address research questions related specifically to
`bioequivalence.
`In the early 19705, bioequivalence
`was evaluated with a basic comparison of the mean
`AUC and Cmax values for the test versus reference
`drug products. That is, if the mean AUC and Cmax val-
`ues for the test product were within 20% of the mean
`values from the reference product, bioequivalence
`was concluded. Other relatively basic criteria (e.g.,
`the 75/75 or 75 / 75—125 rules) have also been used to
`demonstrate bioequivalence. According to these crite-
`ria, if the ratio of the AUC and Cmax of the test product
`versus those of the reference product was within 75%
`to 125% for at least 75% of the subjects, bioequivalence
`could be claimeddalfifl
`In accordance with the conclusions from a 1986
`
`the FDA began to
`FDA Bioequivalence Task Force,
`recommend a more standard approach for in viva
`bioequivalence studies in the early 19905.18 This
`approach is known as average bioequivalence,
`in
`which the statistical analysis of pharmacokinetic
`measurements (e.g., AUC and Cmax) is based on the
`two one-sided test (TOST) procedures to determine if
`the average values from the test product are compara-
`ble to those of the reference product. Using this pro-
`cedure, a 90% confidence interval for the difference of
`
`the pharmacokinetic measurements is calculated on a
`log scale. Bioequivalence is demonstrated if this calcu-
`lated confidence interval falls within the predefined
`bioequivalence limits (i.e., :022 with a log scale or
`0.8 to 1.25 with a regular scale).
`In the late 19905,
`the FDA subsequently recom-
`mended two different approaches.19 These new
`approaches are known as population bioequiva-
`lence, and individual bioequivalence, and each can be
`compared and contrasted to the standard approach
`(i.e., average bioequivalence). From a statistical point
`of view, for example, average bioequivalence focuses
`on comparing the population average values of the
`pharmacokinetic measurements from two product
`groups. By contrast, population bioequivalence and
`individual bioequivalence focus on comparing the
`average values, and their variances as well. These
`variance terms are included in the population and
`individual bioequivalence approaches to reflect the
`possibility that the test and reference drug products
`
`ll. BIOPHARMACEUTICAL AND PHARMACOKINETIC EVALUATIONS OF DRUGS MOLECULES AND DOSAGE FORMS
`
`InnoPharma Exhibit 1023.0007
`
`

`

`
`
`15. BIOAVAILABILITY AND BIOEQUIVALENCE
`
`could differ in some ways other than in their average
`bioavailabilities.
`
`Population bioequivalence assumes an equal distri-
`bution of the test and reference formulation bioavail-
`
`abilities across all subjects in the population, and is
`intended to address the issue of drug prescribability.
`Drug prescribability refers to the clinician’s choice for
`prescribing a particular drug product versus another
`product to a new patient. For example, a clinician can
`choose to prescribe either the brand name drug or any
`number of generic versions that have been shown to
`be bioequivalent. The underlying assumption of drug
`prescribability is that the brand name drug product
`and its generic versions may be used interchangeably
`in terms of general efficacy and safety.
`focuses on
`Individual bioequivalence, however,
`drug switchability, and the assumption of equivari—
`ance of the distributions of the test and reference prod-
`uct bioavailabilities for each individual subject. Drug
`switchability describes the situation when a clinician
`prescribes an alternative drug product (e.g., a generic
`company’s version) to a patient who has already been
`titrated to a steady, therapeutic level of a previously-
`prescribed drug product (e.g., an innovator compa-
`ny’s drug product). Drug switchability is considered
`more critical, and also more clinically relevant, than
`drug prescribability, particularly for patients with
`chronic diseases requiring long-term drug therapy.
`Drug prescribability and drug switchability can be
`assessed by population bioequivalence and individual
`bioequivalence tests, respectively.”21 The population
`bioequivalence approach is not applicable to evalua-
`tions of drug switchability, however, because of a pos-
`sible subject-by-formulation interaction.22
`
`Average Bioequivalence
`
`Average bioequivalence indicates that two products
`may be considered equivalent if the difference in their
`population average value is relatively small, and con-
`tained within a predefined acceptable range. Under
`current regulatory guidance, the lower and upper lim-
`its (6L and 6U, respectively) of an acceptable range are
`defined as :0.22 in the log scale or from 0.8 to 1.25
`in the original scale. The selection of the lower and
`upper limits for equivalence is somewhat arbitrary,
`and is usually attributed to the general clinical obser-
`vation that for most drugs, a 20% change in dose does
`not significantly change the clinical outcome.
`In average bioequivalence,
`the hypotheses to be
`tested include:
`
`Ho:/1,T — HR 3 6L or #7 —;1.R 2 61
`
`HazéL <(/1.T — MR) < (5U
`
`where M and MR are the population averages of the
`pharmacokinetic measurement (e.g., log AUC or log
`Cmax) for the test and reference products, respectively,
`and 6L and 6H are the prespecified lower and upper
`limits, respectively.
`Average bioequivalence is systematically evalu—
`ated through the following set of procedures. First, a
`non-replicated or replicated bioequivalence study is
`conducted, and intensive blood samples are obtained
`from subjects who received either the test or reference
`product. Next, a non-compartmental analysis is per-
`formed to obtain the pharmacokinetic measurements
`(e.g., AUC or Cmax) and these results are log-trans—
`formed. A linear mixed-effect model is then fitted to the
`
`log AUC (or log Cmax), in which subject and period
`effects are eliminated so that the product (or formu-
`lation) effect, pT — MR, can —be estimated. The stand-
`ard error of the estimated product (or formulation)
`effect is estimated. The 90% confidence interval is
`
`then set by applying the standard error, and compar-
`ing it with the predetermined limits. If the 90% con-
`fidence interval falls within the predetermined upper
`and lower limits (i.e., 6L and (SH) bioequivalence can be
`concluded.
`
`For bioequivalence analyses, the confidence inter-
`val approach is considered more plausible than the
`conventional hypothesis test approach. Conventional
`hypothesis tests (e.g., t test or F test), generally test
`the null hypothesis that M — [LR = 0. Once the null
`hypothesis is rejected under a certain preset alpha
`level (e.g., a = 0.05), the type I error is under con-
`trol, and the probability of falsely declaring ineq-
`uivalence
`is
`also thereby controlled. However,
`controlling instead for the error of falsely declaring
`equivalence would actually be more meaningful in
`tests of bioequivalence. This relates to the power func-
`tion in a conventional hypothesis test. Power functions
`cannot be directly tested. However, a confidence inter-
`val can be linked to the power function that is defined
`as the probability that the confidence interval would
`not include an alternative value. Bioequivalence can
`then be concluded at a risk no greater than 10%, once
`the 90% confidence interval has no common points
`with the equivalence limits or the 90% confidence
`interval falls within the equivalence limits.
`Westlake23 initially suggested that a confidence
`interval approach could be applied in bioequivalence
`studies and, when it was first introduced, he proposed
`using a confidence interval centered on the point of
`exact equivalence (i.e., HT —;1,R = 0). Kirkwood,24 on
`the other hand, suggested using a conventional confi-
`dence interval, which is instead centered on the point
`estimate (0T — [1.12). Thus, with Westlake’s approach,
`equivalence is accepted if the probability of equivalence
`
`ll. BIOI‘H

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