Generic Drug Product Development: SPH SPH IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count
Generic Drug Product Development: SPH SPH IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count
Generic Drug
Product Development
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Executive Editor
James Swarbrick
PharmaceuTech, Inc.
Pinehurst, North Carolina
Advisory Board
Generic Drug
Product Development
International Regulatory Requirements
for Bioequivalence
Edited by
Isadore Kanfer
Rhodes University
Grahamstown, South Africa
Leon Shargel
Applied Biopharmaceutics, LLC
Raleigh, North Carolina, USA
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
This book contains information obtained from authentic and highly regarded sources. While all reasonable
efforts have been made to publish reliable data and information, neither the author[s] nor the publisher can
accept any legal responsibility or liability for any errors or omissions that may be made. The publishers wish to
make clear that any views or opinions expressed in this book by individual editors, authors or contributors are
personal to them and do not necessarily reflect the views/opinions of the publishers. The information or guid-
ance contained in this book is intended for use by medical, scientific or health-care professionals and is provided
strictly as a supplement to the medical or other professional’s own judgement, their knowledge of the patient’s
medical history, relevant manufacturer’s instructions and the appropriate best practice guidelines. Because of
the rapid advances in medical science, any information or advice on dosages, procedures or diagnoses should be
independently verified. The reader is strongly urged to consult the drug companies’ printed instructions, and
their websites, before administering any of the drugs recommended in this book. This book does not indicate
whether a particular treatment is appropriate or suitable for a particular individual. Ultimately it is the sole
responsibility of the medical professional to make his or her own professional judgements, so as to advise and
treat patients appropriately. The authors and publishers have also attempted to trace the copyright holders of all
material reproduced in this publication and apologize to copyright holders if permission to publish in this form
has not been obtained. If any copyright material has not been acknowledged please write and let us know so we
may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmit-
ted, or utilized in any form by any electronic, mechanical, or other means, now known or hereafter invented,
including photocopying, microfilming, and recording, or in any information storage or retrieval system, without
written permission from the publishers.
For permission to photocopy or use material electronically from this work, please access www.copyright.com
(http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Dan-
vers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and registration for a
variety of users. For organizations that have been granted a photocopy license by the CCC, a separate system of
payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only
for identification and explanation without intent to infringe.
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com
Preface
CONTENTS
The bioequivalence regulations and requirements of the following countries and
their associated regulatory agencies are included in this book:
vii
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
viii Preface
Preface ix
the implications of their different requirements and approaches for the market
approval of a generic product, particularly with respect to it’s declaration of
interchangeability/switchability.
A list of definitions, abbreviations and symbols has been included where
many of the terms are commonly used in most countries.
Isadore Kanfer
Leon Shargel
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Acknowledgments
The editors wish to express their gratitude to all the authors who have given so
generously of their time and expertise to provide these texts and to the organiza-
tions, for which they work, which have graciously allowed them to do so.
x
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Disclaimer
The contents of this book represent the views of its authors. They do not nec-
essarily reflect the policies or opinions of any national authorities (regulatory
agencies), advisory or regulatory committees, pharmaceutical companies, con-
tract research organizations, consultancy companies, academic institutions, hos-
pitals, etc., with whom the authors may be associated or employed.
xi
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Annexure 1
Definitions, Abbreviations and Symbols
xii
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
site of drug action when administered at the same molar dose under
similar conditions in an appropriately designed study.”
Bioequivalence focuses on the equivalence of release of the
active pharmaceutical ingredient from the pharmaceutical product
and its subsequent absorption into the systemic circulation.
Comparative studies using clinical or pharmacodynamic end
points may also be used to demonstrate bioequivalence.
FDC Fixed-dose combination
A combination of two or more active pharmaceutical ingredients in a
fixed ratio of doses. This term is used generically to mean a particular
combination of active pharmaceutical ingredients irrespective of the
formulation or brand. It may be administered as single entity
products given concurrently or as a finished pharmaceutical product.
FP Final product
A product that has undergone all stages of production, excluding
packaging.
FPP Finished pharmaceutical product
A product that has undergone all stages of production, including
packaging in its final container and labeling.
IPI Inactive pharmaceutical ingredient
A substance or compound that is used in the manufacture of a
pharmaceutical product and does not contribute to the therapeutic
effect of the product, but is intended to enhance the consistency,
appearance, integrity, stability, release characteristics, or other
features of the product.
Usually refers to excipients or other formulation
adjuvants/aids.
MSPP Multisource (generic) pharmaceutical product
Multisource pharmaceutical products are pharmaceutically
equivalent products that may or may not be therapeutically
equivalent or bioequivalent. Multisource pharmaceutical products
that are therapeutically equivalent are interchangeable.
MRDF Modified-release dosage forms
A modified-release dosage form is one for which the drug-release
characteristics of time course and/or location are chosen to
accomplish therapeutic or convenience objectives not offered by
conventional dosage forms such as solutions, ointments, or promptly
dissolving dosage forms.
Delayed-release and extended-release dosage forms are two
types of modified-release dosage forms.
r Delayed-release dosage forms: A delayed-release dosage form is one
that releases a drug(s) at a time other than promptly after admin-
istration.
r Extended-release dosage forms: An extended-release dosage form is
one that allows at least a twofold reduction in dosing frequency
or significant increase in patient compliance or therapeutic per-
formance as compared to that presented as a conventional dosage
form (e.g., as a solution or a prompt drug-releasing, conventional
solid dosage form).
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Contents
Preface . . . . vii
Acknowledgments . . . . x
Disclaimer . . . . xi
Annexure 1: Definitions, Abbreviations and Symbols . . . . xii
Contributors . . . . xx
1. Introduction 1
Isadore Kanfer
2. Australasia 17
C. T. Hung, D. Ren, L. A. Folland, F. C. Lam, Noelyn Anne Hung,
and R. Smart
3. Brazil 46
Margareth R. C. Marques, Sı́lvia Storpirtis, and Márcia Martini Bueno
4. Canada 67
Iain J. McGilveray
6. India 114
Subhash C. Mandal and S. Ravisankar
7. Japan 164
Juichi Riku
xviii
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Contents xix
Index . . . . 301
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Contributors
xx
SPH SPH
IHBK055-FM IHBK055-Kanfer January 8, 2010 19:20 Char Count=
Contributors xxi
1 Introduction
Isadore Kanfer
Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa
INTRODUCTION
The introduction and application of bioequivalence testing as a surrogate indica-
tor of safety and efficacy has had an enormous impact on the development and
formulation of solid oral dosage forms in particular, not only for generic prod-
uct manufacturers but also for the innovator or “brand” companies. Using bioe-
quivalence as a tool to establish therapeutic equivalence between a test (generic)
and a reference product certainly forms the basis for market approval of mul-
tisource drug products, the latter being products marketed by more than one
manufacturer and containing the same active pharmaceutical ingredient (API) in
the same dosage form intended to be administered by the same route of admin-
istration. The reference product is usually the comparator product developed
and marketed by a so-called “innovator” or brand company, where their prod-
uct had been approved on the basis of clinical trials to support claims for safety
and efficacy. It is however a little-known fact that many innovator products that
eventually become available on a particular market were in fact approved on the
basis of a bioequivalence study (1) between the formulation used in the clinical
trials (as reference) and an amended formulation of the same active(s), that is, the
test product. Hence, it is apparent that using bioequivalence methodology and
sound scientifically based acceptance criteria to show therapeutic equivalence
overcomes the need to redo clinical trials that are expensive and time consum-
ing, often taking several years to produce outcomes. As mentioned previously,
it is in the area of solid oral dosage forms where the major benefits of bioequiv-
alence testing occur since plasma drug concentrations can be measured follow-
ing oral administration of a test and reference product in a cross-over fashion
in healthy human subjects. This procedure is feasible and scientifically sound
for drugs, which are intended to be absorbed into the systemic circulation since
the underlying principle is that a link exists between drug concentrations in the
blood and therapeutic effect. However, for other dosage forms, particularly those
intended for other routes of administration and more specifically those which
are not intended to be absorbed into the systemic circulation, generally no such
link exists between drug concentrations in blood and effect. Although some spe-
cific methodologies have been developed and validated, such as the human skin
blanching assay (HSBA) for topical corticosteroid products, to date relatively few
surrogate measures to assess bioequivalence of such dosage forms have been
accepted by regulatory agencies. It is thus apparent that the main objective of
bioequivalence testing is to compare the formulation performance between a test
and reference product with the premise that if bioequivalence is proven, there
1
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
2 Kanfer
a Approved Drug Products with Therapeutic Equivalence Evaluations,” Orange Book, (www.
fda.gov/downloads/Drugs/DevelopmentApprovalProcess/UCM071436.pdf).
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
Introduction 3
BRAZIL
In Brazil, the National Policy for Drug Products published in 1998 created the
National Agency for Sanitary Surveillance (Agencia Nacional de Vigilancia Sani-
taria, ANVISA), which is responsible for the approval of the law and the publica-
tion of the technical guidances for the registration of generic products (5–8). The
law and guidances for the registration of generic products in Brazil are based on
the regulations of countries such as Canada, United States of America, and the
EU. Brazil was the first country in South America to implement the evaluation
of pharmaceutical equivalency and bioequivalence studies for the registration of
generic products.
CANADA
Canadian regulators, under the Health Products and Food Branch (HPFB), Ther-
apeutic Products Directorate (TPD), Health Canada, were the first to apply phar-
macokinetics (PK) to safety and efficacy risk assessment of generic drug products
as a consequence of 1969 amendments to the Patent Act (9) (compulsory licens-
ing). Guidelines [Reports A, B, and C (10–12)] were only published in the 1990s
by an Expert Advisory Committee (EAC), currently referred to as the Scientific
Advisory Committee (SAC). However, Canada is governed as a Confederation
of Provinces and the regulations and guidelines for bioequivalence are federal,
leading to a Notice of Compliance (NOC) to sponsors for marketing in Canada.
Although an application may lead to a Declaration of Bioequivalence for specific
products, the various Canadian provinces and associated Provincial formulary
committees may not accept the Federal decision of bioequivalence (which also
evaluates quality) to be sufficient to list a particular product as interchangeable.
Revision of the Canadian guidelines was mentioned during presentations
at the 2008 and 2009 annual meetings of the Canadian Society of Pharmaceu-
tical Sciences (CSPS) held in Banff during May 2008 and in Toronto during
June 2009 (13).
EUROPEAN UNION
The EU has established requirements which must be met by a generic drug prod-
uct to receive marketing authorization (14). The EU offers four routes for the
registration of generic drug products (15):
1. National Procedure
2. Mutual Recognition Procedure
3. Decentralized Procedure
4. Centralized Procedure.
The national procedure (NP) is strictly limited to medicinal products that
are not authorized in more than one member state and may lead to marketing
authorization of the generic drug product in the concerned member state. The
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
4 Kanfer
mutual recognition procedure (MRP) makes provision for the extension of market-
ing authorizations granted to one member state, the so-called reference member
state (RMS), to one or more member states identified by the applicant. The decen-
tralized procedure (DCP) was implemented from November 2005 where a submis-
sion can be made to each of the member states, where it is intended to obtain a
marketing authorization with a choice of one of them as the RMS. The centralized
procedure (CP) has been in use since 2004 for marketing authorization of medic-
inal products in the EU. Here a single application is introduced for evaluation
that is carried out within the Committee for Medicinal Products for Human Use
(CHMP) of the EMEA and is valid throughout the EU with the same rights and
obligations in each of the member states.
The first European bioequivalence guidelines were published in 1991 (16)
and assessment was based on the principles published in the scientific literature,
FDA guidelines, and the first European guidelines on pharmacokinetic studies
in man (17). In 2001, the EMEA’s CPMP published the current version of the
Note for Guidance on the Investigation of Bioavailability and Bioequivalence
(18). A draft version of revised bioequivalence guidelines, entitled Guideline
on the Investigation of Bioequivalence, was made publicly available in August
2008 on the EMEA Web site and a modified version is due to come into effect
in 2010 (19).
INDIA
Bioequivalence assessment for generic medicines in India was instituted by the
incorporation of Schedule Y of the Drugs and Cosmetics Act in 1988, followed
by subsequent amendments of Schedule Y in 1989 and 2005 (20).
In India, generic medicines are those medicines, which are labeled with
their generic names. There is no separate law for registering generic medicines.
Drugs and drug products are classified as either “new drugs” or drugs other than
new drugs. However, in general “generic drugs” refer to those drugs than are no
longer subject to patent protection and are being marketed by their generic name.
In fact, it’s interesting to note that India only started observing product patents
from January 1, 2005 (21,22).
India has a two tier regulatory system, the Central Drugs Standard Control
Organization (CDSCO) under the Government of India and each State has its
own drug regulatory system having certain powers.
JAPAN
In Japan, data from 2006 indicated that generic drug products accounted for as
little as 16.9% of the market by volume 5.7% by value. In the following year,
the Japanese government announced a specific program to increase the use of
generic products to more than 30% by the year 2012 and a system for generic
substitution was targeted for implementation in April 2008.
Approval to manufacture and market generic drug products in accordance
with the Pharmaceutical Affairs Law in Japan is granted by the Minister of
Health, Labour and Welfare (MHLW). An independent administrative organiza-
tion, the Pharmaceutical and Medical Devices Agency (PMDA), under the aus-
pices of the MHLW, undertakes the review and assessment of bioequivalence
data as part of the “Equivalence and Compliance Review.”
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
Introduction 5
The first guideline for bioequivalence studies for generic dugs was released
in 1971, in which large animals, such as dogs and rabbits, could be used in bioe-
quivalence studies, but humans were not required. Subsequently in 1997, the
MHLW published a revised Guideline “The Guideline for Bioequivalence Stud-
ies of Generic Products 1997 (23) was introduced. Several newer guidelines and
revisions have been published such as the Guideline for Bioequivalence Studies
for Different Strengths of Oral Solid Dosage Forms 2000 (24), Guideline for Bioe-
quivalence Studies for Additional Dosage Forms of Oral Solid Dosage Forms
2001 (25), Guideline for Bioequivalence Studies of Generic Products for Topical
Dermal Application 2003 (26), Guideline for Bioequivalence Studies for Formu-
lation Changes of Oral Solid Dosage Forms 2000 (27), Draft Guideline for Bioe-
quivalence Studies for Changes in Manufacturing of Oral Solid Dosage Forms:
Conventional, Enteric Coated and Prolonged Release Products, 2003 (28), and
Revision of Guideline for Bioequivalence Studies of Generic Products, 2006 (29).
For oral drug products, in Japan, dissolution testing of these dosage forms
is required together with bioequivalence studies and plays an important role in
selecting appropriate subjects for the in vivo study. In fact, it is noteworthy that
the use of dissolution testing for BA/BE in Japan clearly marks a distinct differ-
ence between Japanese BA/BE guidelines and those of most other countries.
SOUTH AFRICA
The Medicines Control Council (MCC) in South Africa is a statutory body that
was established in terms of the Medicines and Related Substances Control Act
(MRSCA), 101 of 1965, to oversee the regulation of medicines in South Africa.
To facilitate the registration process for generic medicines, guidelines have been
prepared to serve as a recommendation to applicants wishing to submit data in
support of the registration of such medicines (30–33).
In the early 2000s, the requirements for the registration and market
approval of generic medicines were published as official guidelines. Prior to
that time, proof of safety and efficacy of generic medicines were based on
requirements described in “official” notices or circulars issued by the MCC. In
many instances, only in vitro dissolution testing was required based upon Cir-
cular 14/95 that was first issued in the early 1990s and subsequently updated
in 1995 (34).
During 2002 (35,36), legislation was introduced to make provision for
generic substitution whereby dispensers of medicines were mandated to inform
patients of the benefits of the substitution for a branded medicine by an inter-
changeable multisource medicine and to recommend accordingly. The final deci-
sion, however, has been left in the hands of the patient.
6 Kanfer
TAIWAN
The Bureau of Pharmaceutical Affairs (BPA) within the Department of Health
(DOH) is responsible for the regulation of medicinal products in Taiwan. In 1984,
the BPA outsourced the drafting of BA/BE guidelines and Taiwan guidelines on
BA/BE for generic medicines were issued in 1987. Generic products are reviewed
and approved within the BPA at the DOH. The applicant needs to certify to the
DOH that the patent in question is not infringed by the generic product. Once
the BPA staff have completed the filing review of the submission and have ver-
ified that the application has met all the necessary regulatory requirements, the
application is then assigned to technical review which focuses on BE data and
chemistry and manufacture quality.
TURKEY
Marketing authorization for medicinal products for humans are issued by the
Ministry of Health (MoH) in Turkey, but the entire procedure is managed by the
General Directorate of Drug and Pharmacy (GDDP). Turkish licensing regula-
tions for all pharmaceutical products, innovator/brand and generics, came into
force on June 30, 2005 (37). This legislation brought Turkish law in line with that
of the EU and covered all aspects of the drug registration procedure and all new
Turkish regulations are intended to be, as far as possible, compatible with those
of the EU. However, bioequivalence became compulsory for a generic drug prod-
uct to receive a marketing license after the publication of a regulation on May 27,
1994 (38). In general, the design and conduct of a BE study should follow Turkish
and/or EU regulations on Good Clinical Practice (GCP).
b The Drug Price Competition and Patent Term Restoration Act of 1984, informally known as
the ”Hatch-Waxman Act” is an amendment to the U.S. Food and Drug Law that established
the Abbreviated New Drug Application (ANDA) as a pathway for approval of generic drug
products.
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
Introduction 7
products (41), which was revised in 1992 (42). The current FDA guidance for
industry was posted in 2003 and updates recommendations for documentation
of bioavailability and bioequivalence required for regulatory submissions (43).
Although this guidance indicates that it provides recommendations for orally
administered drug products, the guidance also applies to nonorally administered
drug products where reliance on systemic exposure measures is suitable to docu-
ment bioavailability/bioequivalence such as for transdermal and some rectal and
nasal products. Several separate guidances have also been published and which
relate to topical dermatologic corticosteroids (44), food effects (45), and guidance
on waivers for in vivo studies for immediate-release solid oral dosage forms on
the basis of the Biopharmaceutics Classification System (BCS) (46). On Friday,
January 16, 2009, “Final rule” on the requirements for submission of Bioequiv-
alence Data was published in the Federal Register (47). Several other guidances
that have been published are given here.
CDER Guidance for Industry: BE Recommendations for Specific Products
(48) and CDER Guidance for Industry: Individual Product Bioequivalence Rec-
ommendations (49). These latter two guidances provide information on specific
drug products relating to the design of BE studies to support ANDAs and infor-
mation on individual products. Another draft, Guidance on the Submission of
Summary Bioequivalence Data for ANDAs (50), relates to the FDA’s require-
ment that ANDA applicants submit data from all BE studies that the applicant
conducts on a drug product formulation including studies that do not demon-
strate bioequivalence of their generic product submitted for approval. This guid-
ance also provides information on the format for summary reports of BE studies
and types of formulations that the FDA considers to be the same drug product
formulation for different dosage forms, based on differences in composition.
The ANDA is submitted to the Office of Generic Drugs (OGD)c and upon
filing acceptance the application is assigned for bioequivalence review, where the
review process assesses the bioequivalence data comparing the generic product
and the reference listed drug (RLD) indicated in the Orange Book (51).
8 Kanfer
d Pharmaceutical alternatives may be a capsule and a tablet containing the same active drug
substance in the same dose strength. However, the U.S. FDA does not allow capsule formu-
lations to be interchanged for tablet formulation of the same drug even if both products are
bioequivalent.
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
Introduction 9
10 Kanfer
Introduction 11
BIOWAIVERS
The main intentions of a biowaiver is to provide a cost-effective approach to
improve the efficiency of drug development and the review process by recom-
mending a strategy for identifying expendable clinical bioequivalence tests.e The
BCS published by Amidon et al. in 1995 (53) finally lead to consideration of
the possibility of waiving in vivo bioequivalence studies for certain immediate-
release drug products in favor of specific comparative in vitro testing to con-
clude bioequivalence without requiring an in vivo bioequivalence study. This
approach is meant to reduce unnecessary in vivo bioequivalence studies and the
use of human subjects but is, however, restricted to noncritical drug substances
in terms of solubility, permeability, and therapeutic range, and to noncritical
e http://www.fda.gov/AboutFDA/CentersOffices/CDER/ucm128219.htm.
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
12 Kanfer
SUMMARY
Most countries appear to have adopted the basic tenets and approaches used by
those countries that pioneered (Canada and United States) the introduction and
application of this particular tool for the assessment of bioequivalence of differ-
ent formulations of the same drug product. Apart from its general use, both by
innovator and generic drug companies, there still appears to be a certain degree
of naivety and ignorance associated with BE testing where the notion persist that
it is specifically a tool for use only in the assessment of generic drug products.
However, some causes for concern still remain, and those relate to issues
of semantics and interpretation of the main objectives such as, for example, the
association between bioequivalence, therapeutic equivalence, and generic sub-
stitution. In particular, statements such as,“may or may not be interchangeable,”
or “may or may not be bioequivalent or therapeutically equivalent,” which appear in
some formal definitions such as the following are disconcerting.
r Note: a product that has been approved based on comparison with a nondo-
mestic comparator product may or may not be interchangeable with currently
marketed domestic products (Ref. 52, 6.5.2, p. 364).
r Multisource pharmaceutical products (52).
Pharmaceutically equivalent or pharmaceutically alternative products that
may or may not be therapeutically equivalent. MPP that are therapeutically
equivalent are interchangeable.
r Pharmaceutical alternatives (52).
Pharmaceutical alternatives deliver the same active moiety by the same route
of administration but are otherwise not pharmaceutically equivalent. They
may or may not be bioequivalent or therapeutically equivalent to the compara-
tor product.
Introduction 13
REFERENCES
1. Benet LZ. Understanding bioequivalence testing. Transplant Proc 1999; 31(suppl
3A):7S–9S.
2. Therapeutic Goods Act 1989—Australian Government Publishing Service, Canberra.
3. Medicines Act 1981 (Reprint 2006) (published under the Authority of the New
Zealand Government).
4. Note for Guidance on the investigation of bioavailability and bioequivalence
(CPMP/EWP/QWP/1401/98). CPMP Guidance—As adopted by the TGA—with
amendment. April 10, 2002. http://www.tga.gov. Accessed May 30, 2009.
5. Health Minister. National Health Surveillance Agency. MS/GM Ordinance no. 3916
of October 30, 1998. Approval of the National Drug Policy. Brazil. Brası́lia, DF; 1998.
http://e-legis.bvs/leisref/public/showAct.php?id=751. Accessed May 30, 2009.
6. Brazil. Law No. 9782 of January 26, 1999. Definition of the National Health
Surveillance System, establishment of the National Health Surveillance Agency,
among other provisions. Brası́lia, DF; 1999. http://e-legis.bvs/leisref/public/
showAct.php?id=182. Accessed May 30, 2009.
7. Brazil. Law N. 9787 of February 10, 1999. Amendment of Act No 6360 deals
with health surveillance and provides for the use of generic names in pharma-
ceutical and other provisions. Brası́lia, DF; 1999. http://e-legis.bvs/leisref/public/
showAct.php?id=245. Accessed May 30, 2009.
8. Brazil. Health Minister. National Health Surveillance Agency. Resolution N. 391 of
August 9, 1999. Technical regulation for generic drugs. Brası́lia, DF; 1999. http://
e-legis.bvs/leisref/public/showAct.php?id=251. Accessed May 30, 2009.
9. Bill C-102 An Act to Amend the Patent Act, the Trademarks Act and the Food and
Drugs Act. See www. curc.clc-ctc.ca/lang. Accessed May 30, 2009.
10. Guidance for Industry. Conduct and Analysis of Bioavailability and Bioequivalence
Studies – Part A: Oral Dosage Formulations Used for Systemic Effects. Health
Canada 1992. http://www.hc-sc.gc.ca/dhp-mps/alt formats/hpfb-dgpsa/pdf/
prodpharma/bio-a e.pdf. Accessed May 30, 2009.
11. Guidance for Industry. Conduct and Analysis of Bioavailability and Bioequivalence
Studies – Part B: Oral Modified Release Formulations, Health Canada 1996 http://
www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-demande/guide-ld/bio/bio-b-eng
.php. Accessed May 30, 2009.
12. Expert Advisory Committee on Bioavailability, Health Protection Branch, Decem-
ber 1992. Report C: Report on bioavailability of oral dosage formulations, not
in modified-release form, of drugs used for systemic effects, having complicated
or variable pharmacokinetics. Health Canada, 1992. http://www.hc-sc.gc.ca/dhp-
mps/prodpharma/applic-demande/guide-ld/bio/biorepc biorapc-eng.php. Acces-
sed May 30, 2009.
13. Presentations by Eric Ormsby (Health Canada) to the Canadian Society of Phar-
maceutical Scientists Annual Meetings, (i)Banff, Alberta, May 24, 2008 (ii)Toronto,
Ontario, June 6, 2009.
14. Chen M-L, Shah V, Patnaik R, et al. Bioavailability and bioequivalence: An FDA reg-
ulatory overview. Pharm Res 2001; 18:1645–1650.
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
14 Kanfer
15. EC Notice to Applicants, volume 2A: Procedures for marketing authorisation. The
Rules Governing Medicinal Products in the European Union, Chapter 1: Marketing
Authorisation, November 2005. http://ec.europa.eu/enterprise/pharmaceuticals/
eudralex/vol-2/a/vol2a chap1 2005–11.pdf. Accessed May 30, 2009.
16. CPMP Note for Guidance on the Investigation of Bioavailability and Bioequivalence,
Brussels, December 1991.
17. Pharmacokinetic Studies in Man, 1987. http://www.emea.europa.eu/pdfs/human/
ewp/3cc3aen.pdf. Accessed May 30, 2009.
18. EMEA Note for Guidance on the Investigation of Bioavailability and Bioequiva-
lence, London, July 2001, CPMP/EWP/QWP/EMEA. http://www.emea.europa.eu/
pdfs/human/qwp/140198enfin.pdf. Accessed May 30, 2009.
19. EMEA Guideline on the Investigation of Bioequivalence, EMEA, London, July
24, 2008, CPMP/EWP/QWP/1401/98 Rev. 1. http://www.emea.europa.eu/pdfs/
human/qwp/140198enrev1.pdf. Accessed May 30, 2009.
20. The Drugs and Cosmetics Act 1940 and The Drugs and Cosmetics Rules 1945, Min-
istry of Health and Family Welfare, Government of India, 2005:425–459.
21. The Patents (Amendment) Act 2005, The Gazette of India: Extraordinary, Part II-Sec.
I, 1–18, 2005.
22. The Patents (Amendment) Rules, 2005, The Gazette of India: Extraordinary, Part II-
Sec. 3 (ii), 60–108, 2004.
23. Ministry of Health, Labour and Welfare. Guideline for Bioequivalence Stud-
ies of Generic Products, 1997. http://www.nihs.go.jp/drug/be-guide(e)/Generic/
be97E.html. Accessed May 30, 2009.
24. Ministry of Health, Labour and Welfare. Guideline for Bioequivalence Studies for
Different Strengths of Oral Solid Dosage Forms, 2000. http://www.nihs.go.jp/drug/
be-guide(e)/strength/strength.html. Accessed May 30, 2009.
25. Ministry of Health, Labor and Welfare. Guideline for Bioequivalence Studies for
Additional Dosage Forms of Oral Solid Dosage Forms, 2001.
26. Ministry of Health, Labor and Welfare. Guideline for Bioequivalence Studies of
Generic Products for Topical Dermal Application, 2003. http://www.nihs.go.jp/
drug/DrugDiv-E.html. Accessed May 31, 2009.
27. Ministry of Health, Labor and Welfare. Guideline for Formulation Changes of Oral
Solid Dosage Forms, 2000. http://www.nihs.go.jp/drug/DrugDiv-E.html. Accessed
May 31, 2009.
28. Ministry of Health, Labor and Welfare. Draft Guideline for Bioequivalence Studies for
Changes in Manufacturing of Oral Solid Dosage Forms: Conventional, Enteric Coated
and Prolonged Release Products.
29. Ministry of Health, Labour and Welfare. Revision of Guideline for Bioequivalence
Studies of Generic Products, 2006.
30. Pharmaceutical and Analytical: Medicines Control Council, Department of Health,
2.02 P&A, Jun07, v2, 2007. http://www.mccza.com/showdocument.asp?Cat=17&
Desc=Guidelines%20-%20Human%20Medicines. Accessed April 29, 2009.
31. BIOSTUDIES: Medicines Control Council, Department of Health, 2.06 Biostud-
ies, Jun07, v2, 2007. http://www.mccza.com/showdocument.asp?Cat=17&Desc=
Guidelines%20-%20Human%20Medicines. Accessed April 29, 2009.
32. DISSOLUTION: Medicines Control Council, Department of Health, 2.07 Dissolu-
tion, Jun07, v2, 2007. http://www.mccza.com/showdocument.asp?Cat=17&Desc=
Guidelines%20-%20Human%20Medicines – Accessed 29 April 2009
33. Generic Substitution: Medicines Control Council, Department of Health, December
2003. http://www.mccza.com/showdocument.asp?Cat=17&Desc=Guidelines%20-
%20Human%20Medicines – Accessed 29 April 2009
34. Circular 14/95, Data Required as Evidence of Efficacy, Annexure 13, Medicines Con-
trol Council, Department of Health, October, 2, 1995
35. Medicines and Related Substances Control Act, 1965 (Act No. 101 of 1965) as amended
by Act No. 90 of 1997 and Act No. 59 of 2002. http://www.mccza.com. Accessed April
29, 2009.
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
Introduction 15
36. Medicines and Related Substances Control Act, 1965 (Act No. 101 of 1965) as amended
by Act No. 59 of 2002. http://www.mccza.com. Accessed April 29, 2009.
37. Regulation on Licensing for Medicinal Products for Human Use, Official Gazetta No:
25725/19 January 2005
38. Regulation on the Evaluation of Bioavailability and Bioequivalence of Medici-
nal Products Official Gazetta of Turkey No: 21942. http://rega.basbakanlik.gov.tr/.
Accessed May 31, 2009.
39. Conner DP, Davit BM. Bioequivalence and drug product assessment, in vivo. In:
Shargel L, Kanfer I, eds. Generic Drug Product Development: Solid Oral Dosage
Forms. New York: Marcel Dekker, 2005:229.
40. Office of Technology Assessment. Drug Bioequivalence. A Report of the Office of
Technology Assessment Drug Bioequivalence Study Panel. Washington, DC: US Gov-
ernment Printing Office, 1974.
41. 42 Fed Regist 1648, Jan. 7, 1977.
42. 57 Fed Regist 17950, Apr. 28, 1992.
43. US Dept of Health and Human Services, Food and drug Administration, Center
for Drug Evaluation and Research. Guidance for Industry. Bioavailability and Bioe-
quivalence Studies for Orally Administered Drug Products—General Considerations.
March 19, 2003.
44. US Dept of Health and Human Services, Food and Drug Administration, Center for
Drug Evaluation and Research. Guidance for Industry: Topical Dermatological Corti-
costeroids: In Vivo Bioequivalence. March 6, 1998.
45. US Dept of Health and Human Services, Food and Drug Administration, Center for
Drug Evaluation and Research. Draft Guidance for Industry: Food—Effect Bioavail-
ability and Bioequivalence Studies. December 2002.
46. US Dept of Health and Human Services, Food and Drug Administration, Center for
Drug Evaluation and Research. Guidance for Industry: Waiver of In Vivo Bioavail-
ability and Bioequivalence Studies for Immediate-Release Solid Oral Dosage Forms
Based on a Biopharmaceutics Classification System. August 31, 2000.
47. US Dept of Health and Human Services, Food and Drug Administration, 21 CFR Parts
314 and 320, /Vol. 74, No. 11 //Rules and Regulations, [Docket No. FDA–2003–N–
0209] (Formerly Docket No. 2003N–0341) RIN 0910–AC23, Requirements for Submis-
sion of Bioequivalence Data; Final Rule
48. US Dept of Health and Human Services, Food and Drug Administration,
Center for Drug Evaluation and Research Guidance for Industry: BE Rec-
ommendations for Specific Products. http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/ucm072872.pdf, Accessed
June 6, 2009
49. US Dept of Health and Human Services, Food and Drug Administration,
Center for Drug Evaluation and Research Guidance for Industry: Individual
Product Bioequivalence Recommendations. http://www.fda.gov/cder/guidance/
bioequivalence/default.htm. Accessed May 31, 2009.
50. US Dept of Health and Human Services, Food and Drug Administration, Center
for Drug Evaluation and Research Draft Guidance for Industry: Submission of
Summary Bioequivalence Data for ANDAs. http://www.fda.gov/downloads/
Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM134846.pdf.
Accessed June 10, 2009.
51. Approved Drug Products with Therapeutics Equivalence Evaluations, Electronic
Orange Book. Approved Drug Products with Therapeutic Equivalence Evaluations,
Current through April 2009, Department of Health and Human Services, Public
Health Service, Food and drug Administration, Center for Drug Evaluation and
Research, Office of Information Technology, Division of Data Management and Ser-
vices. http://www.accessdata.fda.gov/scripts/cder/ob/default.cfm. Accessed June
10, 2009.
52. Multisource (Generic) Pharmaceutical Products: Guidelines on registration require-
ments to establish interchangeability. In: WHO Expert Committee on Specifications
SPH SPH
IHBK055-01 IHBK055-Kanfer December 28, 2009 14:19 Char Count=
16 Kanfer
2 Australasia
C. T. Hung, D. Ren, L. A. Folland, and F. C. Lam
Zenith Technology Ltd, Dunedin, New Zealand
R. Smart
Douglas Pharmaceuticals, Auckland, New Zealand
INTRODUCTION
History
In Australia, as in many other countries, registration of medicines prior to their
sale became a legal requirement soon after the problems with thalidomide (1)
were identified. At present, the registration of medicines is controlled by the
Therapeutic Goods Act 1989 (2). Under this Act, all medicines sold across state
borders, prescription medicines and any nonprescription medicines intended for
inclusion in the Pharmaceutical Benefits Schedule (PBS), cannot be sold until
they are either registered or listed on the Australian Register of Therapeutic
Goods (ARTG) (www.tga.govt.au). Responsibility for inclusion or otherwise of
medicines on the ARTG rests with the Therapeutic Goods Administration (TGA)
based in Canberra. Prior to the Therapeutic Goods Act 1989, only prescrip-
tion medicines and nonprescription medicines intended for inclusion in the PBS
were evaluated. Nonprescription medicines not intended for inclusion in the PBS
and medicines manufactured and sold within a single state or territory did not
need to receive formal regulatory approval before being sold except in some
states such as the state of Victoria, where registration dubbed “Reg. Vic” by the
Victorian Ministry of Health was required. Hence, in most states, no controls
on the quality, safety, or efficacy of such medicines were enforced. “Reg. Vic”
requirements related to good manufacturing practices (GMP) compliance of the
finished product manufacturer along with evaluation of the quality and labeling
of the finished product. With the coming into effect of the Therapeutic Goods
Act 1989 on the January 15, 1991, these “Reg.Vic” requirements developed some
time later into the OTC compliance branch of the TGA. From July 1, 2006, it
was intended that all medicines, irrespective of scheduling, site of manufacture
or distribution receive regulatory approval from a proposed Trans-Tasman Joint
Regulatory Authority before being permitted for sale. The Joint Agency was to
have responsibility for control of medicines in both Australia and New Zealand
such that both countries were to be viewed as a single regulatory market, with-
out being a single sales market. However in July 2007, the Labor party in the
New Zealand Government at that time, failed to achieve sufficient votes to rat-
ify the legislation designed to give authority to the agency. Establishment of the
17
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
18 Hung et al.
Joint Agency has, therefore been postponed and no indication is presently avail-
able as to when such an agency will come into being. Therefore until such time
as negotiations recommence, the status quo applies with the Therapeutic Goods
Act 1989 being the applicable legislative authority. Continuation of registration
or listing on the ARTG requires payment of an annual fee and no evaluation
occurs with such payment. In relation to the sale of generic medicines, substitu-
tion, at the discretion of the dispensing pharmacist or at the request of the end
user, on prescription is legal in Australia.
In New Zealand, as in Australia, government approval prior to the sale of a
medicine also became a legal requirement soon after the problems with thalido-
mide (1) were identified. New Zealand, however, does not have either a regis-
tration or licensing system. Instead, companies who wish to market medicines
in New Zealand must apply for “Consent to Distribute” (Medicines Act 1981,
Section 20) (3) and cannot begin to market the medicine until such consent is
notified in the New Zealand Gazette. Responsibility for recommending “Consent
to Distribute” rests with Medsafe, a business unit of the New Zealand Ministry
of Health based in Wellington (www.medsafe.govt.nz). Prior to the Medicines
Act 1981 coming into force in 1984, Government approval of medicines was con-
trolled by the Food and Drug Act 1969. Unlike Australia, as New Zealand is a
single country rather than a federation, all medicines irrespective of scheduling,
site of manufacture, or distribution must have “Consent to Distribute” before
sales can commence. “Consent to Distribute,” once notified, remains in force
until 5 years after either the date of the last sale of the product by the company
responsible for placing the product on the market or the date of the last regu-
latory activity such as submission and approval of a Changed Medicine Notifi-
cation, the equivalent of a variation in other regulatory systems. No annual fees
apply. In relation to the sale of generic medicines, substitution on prescription
may only occur where the doctor either writes on the prescription that substitu-
tion of another brand of the same active ingredient is allowed or the prescription
is written using the generic name only of the intended medicine.
In both Australia and New Zealand, medicines may be scheduled as either
prescription medicines, pharmacist medicines, pharmacy only medicines, or gen-
eral sale medicines. In Australia, the scheduling is performed by the Subcommit-
tee on the Uniform Scheduling of Drugs and Poisons (SUSDP), while in New
Zealand, scheduling is the responsibility of an advisory committee to the min-
ister of health known as the Medicines Classification Committee (MCC). The
SUSDP and MCC have worked together closely in the past few years to promote
harmonization of the schedules in each country.
Australasia 19
1. Letter of application
2. Comprehensive table of contents
3. Application form: The template for this form can be downloaded from the
TGA Web site.
4. Australian labeling and packaging
5. Information about the experts
6. Specific requirements for different types of application
7. Drug and Plasma Master Files and Certificates of Suitability of Monographs
of the European Pharmacopoeia: Where products involve manipulation
of blood at some stage, applications must be accompanied by a file, the
“Plasma Master File,” describing that manipulation and the controls
applied. For medicines, the Master file in Module 3.2.S of the application
should include either a full description of the synthesis and controls applied
in manufacturing the active pharmaceutical ingredient (API) as is usual
for applications for new chemical entities (NCEs) or for generics, either
a current Certificate of Suitability issued by the European Directorate for
the Quality of Medicines (www.edqm.eu) or a Drug Master File describing
the synthesis and controls applied in the manufacture of the API. The
information must be accompanied by a specific authorization to allow the
TGA to access the information provided by the API manufacturer on behalf
of the applicant.
8. Good Manufacturing Practice: The TGA requires both the API and finished
product manufacturers located outside Australia to comply with the same
GMP standards as manufacturers resident in Australia. The evidence of
such compliance that is accepted by the TGA is detailed in the 16th edi-
tion of the Guidance on the GMP Clearance of Overseas Manufacturers (6)
available on the TGA Web site.
9. Meetings: A meeting can be arranged between the TGA and an applicant to
discuss and resolve possible issues that are important for a marketing appli-
cation or that may arise during evaluation of an application. The intention
is to reduce barriers to approval of an application
10. Individual patient data: In Australia, individual patient data are not
required to be included as part of a product application except for
the plasma/blood/serum or urine concentrations and derived data from
bioavailability studies. Such data should however, be held by the applicant
in a form suitable for submission in the European Union (EU) or the United
States and be able to be submitted to the TGA on request.
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
20 Hung et al.
11. Overseas regulatory status: The TGA requires applicants to provide a list
of overseas countries where the product has been submitted, the product
information for those countries, any differences between the applications
submitted in those countries and a statement about the regulatory status in
Canada or the United States.
12. Summary of biopharmaceutical studies (defined as bioequivalence and/or
bioavailability studies): Where an application contains biopharmaceutical
studies, the application should include a summary of the study. The appro-
priate template for the summary can be downloaded from the TGA Web site
and covers details such as study design, analytical validation, and pharma-
cokinetic results obtained. If no biopharmaceutical study is included in the
application, justification for not providing such data is required. The infor-
mation to be included can be found in Appendix 15 of the ARGPM.
13. Pediatric development program
Besides the above information there are three additional annexures to be
completed:
1. Environmental risk for non-GMOs (genetically modified organisms) contain-
ing medicines.
2. Antibiotic Resistance Data.
3. Overseas Evaluation Reports.
An application in Australia for a generic product may be classified as either
a Category 1 or a Category 2 application. A Category 1 application is an applica-
tion for a generic product that is not supported by copies of evaluation reports
from two or more overseas regulatory authorities. The Therapeutic Goods Act
1989 requires that the TGA complete the evaluation of a Category 1 application
within 255 working days otherwise the remaining 25% of the evaluation fee is
not paid to the TGA. Where an application for a generic product is supported by
two or more independent evaluation reports of Module 3 data from overseas reg-
ulatory authorities, the application is a Category 2 application, and must be eval-
uated within 120 working days. The TGA currently accepts evaluation reports
from Canada, Sweden, the Netherlands, the United Kingdom, and the United
States. The evaluation reports should be included in Annex 3 to Module 1.
In New Zealand, there is no formal listing of the requirements for inclusion
in Module 1. In general however, the following information should be included.
1. Application form: The same form is used irrespective of whether or not the
medicine is a generic or NCE. Appendices to this form include evidence of
compliance with GMP for the active ingredient manufacturer, finished prod-
uct manufacturer and finished product packer.
2. Generic medicine check list: This is a unique form that must be completed
by an applicant requesting approval of a generic medicine, but not by an
applicant requesting approval of a NCE.
3. Comprehensive table of contents for the whole application.
4. Data sheet: New Zealand has a specific format for presentation of qual-
ity, safety, and efficacy information to health professionals; however, New
Zealand will also accept such information in the form of an SPC (summary of
product characteristics) or Australian Product Information. If a New Zealand
specific data sheet is prepared, it must be accompanied by a completed data
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 21
sheet checklist and declaration. The format of the data sheet, checklist, and
declaration may be found in the New Zealand Regulatory Guidelines for
Medicines (NZRGM) (7) (www.medsafe.govt.nz).
5. Proposed labeling: The requirement is that it should be in the form of either
finished label(s) or finished artwork ready for printing the label(s) or as draft
artwork showing design, color, and wording proposed to be used on the label
for each dose form, strength and pack size. It is emphasized that in New
Zealand, the term “label” refers to the words and designs that are attached to
or part of the container in which the medicine is packed. It does not include
the data sheet or consumer medicine information (CMI).
6. Consumer Medicine Information: The provision of CMI is not a mandatory
requirement; however, it is recommended and if prepared it must be accom-
panied by a declaration of content. The proposed format for CMI and the
declaration to be completed may be found in the New Zealand Guide to the
Registration of Medicines.
22 Hung et al.
BIOEQUIVALENCE REQUIREMENTS
New Zealand
In New Zealand the current system was developed largely in response to the
Cartwright Inquiry, which was concluded in 1988 (11). This inquiry formed the
basis of an independent ethical review process. Further refinements occurred as
a result of the Gisborne Cervical Screening Inquiry in 2001 (12), which recom-
mended that the current process for ethical review be re-evaluated.
The primary role of all ethics committees is the protection of the rights,
health, and well-being of consumers and research participants. Ethics commit-
tee approval is required for all research involving human participants (whether
health or disability support services consumers, healthy volunteers, or members
of the community at large) and where the research falls under one of the spec-
ified categories for matters requiring ethical review. Ethics committees consider
whether the research meets ethical guidelines and considers such matters as to
how participants are selected for the trial and whether they are provided with
enough information to enable them to make an informed decision about partici-
pating. An ethics review is required for each study.
For human ethics, the following committees have been established in
statute, under the New Zealand Public Health and Disability Act 2000:
r The Health Research Council Ethics Committee
r The National Advisory Committee on Health and Disability Support Services
Ethics
r The Ethics Committee on Assisted Reproductive Technology
r Six Regional Health and Disability Ethics Committees and the Multiregion
Ethics Committee
The regional ethics committees consider applications for research that is
to be carried out entirely within just one of New Zealand’s four ethics commit-
tee regions. The multiregion ethics committee considers applications for research
that is carried out in more than one of the ethics committee regions.
Ethics committees are also formed by other organizations and gain accred-
itation through the Health Research Council Ethics Committee (HRCEC). These
include institutional ethics committees (e.g., Universities) and private sector
ethics committees (e.g., Zenith Biomedical Ethics Committee). These committees
may have restrictions upon them as to what type of research they can review
(e.g., healthy volunteer studies only). Any research, however, that involves the
use of patients in the health sector has to be reviewed and approved by a health
and disability ethics committee (or the Multiregion ethics committee if the pro-
posal covers more than one region).
The accreditation of ethics committees by the HRCEC is a formal process.
Every accredited ethics committee is required to provide an annual report plus
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 23
other relevant information as required in the HRC Guidelines for Ethics Committee
Accreditation (13). All accredited ethics committees are required to provide inde-
pendent, competent and timely review of research studies. All members of the
ethics committee are required to be independent, having no connection with any
research proposals that are reviewed, which may cause a conflict of interest.
Ethics committees generally operate in accordance with the ministry of
health’s Operational Standard for Ethics Committees but can also have their own
operational procedures. There is generally a turnaround time of 1 to 2 months
from the ethics committee submission date to obtain full approval.
All applications for research proposals to accredited ethics committees are
completed by way of a formal application process although the exact require-
ments can vary between committees. A requirement of all applications however
is the submission of the study using the National Ethics Application Form.
It is the ethics committee’s responsibility to ensure that adequate compen-
sation provisions are in place for injuries suffered by participants in a clinical
trial. Trials that are sponsored by a pharmaceutical company principally for the
benefit of the manufacturer or distributor are not covered by the Accident Com-
pensation Corporation (ACC). Compensation is provided by the pharmaceuti-
cal company to the extent described in the Researched Medicines Industry Guide-
line (RMI) (14). The “no-fault” principle forms the basis of these guidelines and
the sponsor company should pay compensation to participants suffering bodily
injury in accordance with these guidelines.
A separate approval process is required for clinical trials involving new and
unregistered medicine formulations. The Ministry of Health’s Standing Commit-
tee on Therapeutic Trials (SCOTT) reviews this research in parallel with an ethics
committee application. SCOTT will initially issue a “recommended for approval”
letter, which means it will give final approval once the ethics committee approval
has been obtained. The SCOTT approval process takes a maximum of 45 days
but is generally less. Final approval only takes a few days once ethics committee
approval has been given.
The cost for each SCOTT application is approximately NZ $10,000.
Australia
In Australia, clinical trials of medicines and devices are subject to government
regulations that are administered by the (TGA). Approval for a clinical trial is
gained through a Human Research Ethics Committee (HREC). Only ethics com-
mittees that are constituted and operate in accordance with the National Health
and Medical Research Council’s (NHMRC) National Statement on Ethical Conduct
in Research Involving Humans can approve a clinical trial. HRECs must follow
the guidelines outlined in the National Statement regarding their composition,
appointment of members and various aspects of their operational procedures.
The terms of reference and working procedures of each ethics committee must
be documented. All HRECs must also report annually to the NHMRC through
its principal committee, the Australian Health Ethics Committee. Clinical trials
must be approved by an institutional HREC with jurisdiction at the site where
the study is to be conducted (e.g., hospital, medical center, etc.). In addition,
there is a private ethics committee registered with the NHMRC that can be used
for some sites.
There are two schemes under which clinical trials involving therapeutic
goods may be conducted, the Clinical Trial Notification Scheme (CTN) and the
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
24 Hung et al.
Clinical Trial Exemption (CTX) Scheme. The CTN scheme is a notification scheme
whilst the CTX scheme is an approval process. The vast majority of clinical trials
that are conducted in Australia are approved through the CTN process.
The CTN scheme involves the protocol being submitted directly to the
HREC who is then responsible for assessing the scientific validity and safety of
the project. The TGA does not undertake any review of the study but the sponsor
company is required to submit the CTN form to the TGA along with the appro-
priate fee (presently AU $240) once ethical approval has been granted.
The CTX scheme is intended to assist HRECs when technical, scientific, or
medical data are lacking or where further advice is required. Under this scheme
the sponsor company submits data to the TGA for evaluation and comment. The
CTX application can be made under a 50- or 30-day review and has associated
costs that are substantially higher than a CTN review. Under the CTX scheme,
the TGA will focus primarily on the study in relation to safety issues. An HREC
must also approve the study and the sponsor must wait for written approval
from the TGA before they are permitted to commence their research. All CTN
and CTX trials must have an Australian sponsor.
Compensation for participants for person injury must be provided by the
sponsor company in a form no less favorable than the current version titled
Medicines Australia Form of Indemnity for Clinical Trials (15).
Although clinical trials conducted in New Zealand and Australia are not
required to be registered, registration is recommended. In May 2006, the World
Health Organization (WHO) recommended that all international clinical trials
should be registered. Registration can be completed through the Australian New
Zealand Clinical Trials Registry (www.anzctr.org.au).
There is an intention to establish a Joint Agency that will replace the TGA
and the New Zealand Medicines and Medical Devices Safety Authority (Med-
safe) and be accountable to the Australian and New Zealand Governments.
Participant Selection
Studies should normally be performed using healthy volunteers with the
inclusion/exclusion criteria clearly outlined in the study protocol. Participants
may belong to either sex, with any risk to woman of childbearing potential
considered on an individual basis.
Generally participants should be aged 18 to 55 years and have a weight
within the normal range according to body mass index (BMI) or other appro-
priate accepted weight range indicators (e.g., Metropolitan Life Tables). In New
Zealand and Australia, where the population is multicultural, no restrictions are
placed on the selection of race when conducting studies.
Genetic Phenotyping
Although to date, phenotyping of participants has, to the best of our knowledge,
not been requested by either Medsafe or the TGA, it is worth considering
specifically for parallel design studies where the medicine is known to be
subject to major genetic polymorphism. Phenotyping would allow fast and
slow metabolizers to be evenly distributed in the two groups of participants.
Genetic phenotyping may also be considered for multiple dosing to steady state,
where slow metabolizers are known to present a significantly longer t1/2 of the
active compound. Under these circumstances slow metabolizers should not be
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 25
included in the study since the dosing period would have to be prolonged to
accommodate their inclusion.
Informed Consent
In obtaining and documenting informed consent, investigators should adhere
to GCP (good clinical practice) and the Declaration of Helsinki. Prior to com-
mencement of any study, written approval of (i) information for participants and
(ii) consent to participate documentation should be obtained from an indepen-
dent ethics committee (IEC). Any amendments to this documentation, which
are relevant to participant consent, should be approved by the IEC and revised
copies provided to the participants. The language used in this documentation
should be in lay terms, both practical and understandable to participants.
Participants must give their consent willingly and without coercion or
undue influence from the investigator or study staff. Prior to obtaining con-
sent from any participant the investigator must ensure that each participant is
made aware of the procedures to be carried out during the study. Participants
are required to attend a meeting at which the information for participants form
is read aloud. Participants are encouraged to ask questions relating to the study
medicine, possible side effects, and study procedures.
Prior to informed consent being obtained, subjects are given time to inquire
about details of the study and to decide whether or not to participate. A trial
physician, listed in the study protocol, must be present at each meeting to answer
any medical questions that participants may have and to witness that informed
consent was given freely by each participant. Written informed consent should
be signed and personally dated by the participant and trial physician as well as
an independent witness.
Informed consent should be filed as part of each individual participant
study record.
Participant Screening
The screening and consent of volunteers involves many challenges. In the case of
healthy volunteers, it involves both medical and psychological aspects. Most par-
ticipants with a health science background will request or appreciate knowing
their laboratory results. The most frequently requested result in our experience
is blood type, but obviously this is not a routinely performed test for studies.
26 Hung et al.
examination thereof, which must be respected. Food restrictions are required for
participants from Hindu, Jewish, or Islamic faith, for example.
Medical History
A full medical history is essential and a list of medical conditions forms the basis
of screening. It acts as an aide-memoir for the healthy volunteer who may ignore
or minimize previous illnesses, and can be completed in a relaxed environment
before meeting the examining physician.
Asthma is a commonly experienced condition in childhood, which may
extend into young adulthood, and which a participant will often deny or regard
as a “nonillness,” especially if it is controlled by intermittent inhaler use and
avoidance of precipitating factors. At least a 5-year period free of inhaler use
or symptoms to confirm the participant is suitable for inclusion in a study is
required for study participation.
The medical history should include previous hospital admissions, allergies,
adverse reactions, drug reactions, and complications with anesthesia, in addi-
tion to family medical history. The examining physician also has the opportunity
to assess the participants understanding of the Protocol and procedures, under-
standing of English, and answer any further questions about the study. A mul-
tilingual staff and access to independent interpreters are essential in some cases
to ensure informed consent, understanding of the procedures, and cooperation
with the study requirements.
Provocation Screening
In some instances, especially antibiotics, where the risk of anaphylaxis is higher
than with other drugs, skin testing is performed. The test drug is placed on bro-
ken skin and the response observed and recorded immediately and over the
ensuing days. A full medical response must be in place to treat anaphylaxis. Usu-
ally, any rash is mild and resolves without need for treatment.
Physical Examination
Blood pressure (supine and sitting), height, and weight are recorded. A rise in
systolic blood pressure to about 140 mm Hg is typical for some participants, who
promptly return to a lower pressure when relaxed. Occasionally, a high blood
pressure is found, particularly in older participants (>50 years), females on the
oral contraceptive, and overweight male participants who are then referred to
their family practitioner. Frequently, in the young adult, blood pressure is less
than 100 mm Hg systolic, and this becomes a concern for studies involving blood
pressure lowering drugs. A systolic pressure around 90 mm Hg should exclude
participants from blood pressure lowering studies.
The routine physical examination involves the visual assessment of skin
features (scars, wounds, body fat, peripheral edema) respiratory and cardiac aus-
cultation, abdominal examination, and lymph node examination.
The exclusion of cardiac valve abnormalities is particularly important as
the skin barrier is breached multiple times for venipuncture and intravenous (IV)
line access. An enlarged spleen may be asymptomatic, as may be a cirrhotic liver
or enlarged lymph node.
The forearm skin should be checked for scarring, especially from burns,
which might preclude suitable IV line access. Long forgotten abdominal scars
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 27
may also indicate previous cholecystectomy (perhaps high lipid levels) or the
recent onset of numerous seborrheic keratoses (Leser–Trélat sign of internal
malignancy).
Electrocardiograph
A 12-lead electrocardiograph (ECG) is useful for determining the risk of sudden
cardiac death from prolonged QT interval, short PR interval, or cardiac arrhyth-
mias, for which a participant will be excluded from a study. A copy of the ECG
will be given to the participant along with a referral letter to the family medical
practitioner for further counseling.
In some instances the “stress” of the screening process will be associated
with supraventricular ectopic events, for which a repeat ECG the following day
is recommended.
Laboratory Tests
Medical screening that includes basic hematology, biochemistry, pregnancy tests,
and, drugs of abuse, is designed not only to identify potential medical problems
but also to provide a baseline for possible adverse events.
A hematology screen including full blood count, red cell indices, and ESR
is extremely useful. For example, the young iron deficient vegetarian female par-
ticipant, with a mild anemia may not demonstrate any of the more obvious clin-
ical features of anemia such as pallor, glossitis, stomatitis, or heart failure, but
on prompting will often admit to nonspecific lethargy and weakness. In the pro-
cess of venipuncture during intensive sampling over a short study period, the
hemoglobin may decrease by 10 to 15 g/dL. This can be rapid enough to produce
nonspecific symptoms such as lethargy. Iron replacement therapy may be pre-
scribed or dietary advice given. Also, participants should be within the recom-
mended hemoglobin range to avoid anemia from borderline iron stores (assessed
with a ferritin measurement).
A platelet count is useful to identify idiopathic thrombocytopenic purpura
in the undiagnosed participant. As this is a relatively common disorder in young
or middle-aged adults, medical screening is likely to identify such problems.
Although the ESR is a nonspecific test it has some value if a particularly
high value is obtained (e.g., >100 mm/hr). In the early nonspecific phases of
collagen vascular disorders and malignancy (especially mediastinal Hodgkin’s
lymphoma and multiple myeloma), it may provide an early indicator. While not
diagnostic, it invites referral to the family medical practitioner and further test-
ing, and exclusion from the study.
Biochemistry including plasma sodium and potassium levels provides a
nonspecific overview of homeostatic mechanisms that include renal and nonre-
nal causes of potential abnormalities. Most commonly a raised potassium result
reflects delayed processing or refrigeration of the specimen. Many drugs likely
to be studied (e.g., angiotensin-converting enzyme inhibitors) also run the risk
of hyperkalemia, and normal baseline levels are useful in those instances. In
rare instances the bulimic anorexic participant will present with hypokalemia,
in addition to other factors such as BMI should be helpful.
A screen of liver enzymes can produce small elevations in aspar-
tate aminotransferase (AST), alanine aminotransferase (ALT), or gamma-
glutamyltransferase (gGTP), which can rapidly disappear on repeat testing
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
28 Hung et al.
within days of the initial test. If ALT and/or gGTP are raised, in our experience a
history of alcohol intake 1 or 2 days previously is commonly elicited. Very occa-
sionally the mean red cell hemoglobin is also raised implicating more chronic
use (or abuse) of alcohol. A raised AST level may reflect skeletal muscle derived
enzyme following exercise and this is readily resolved by creatine kinase mea-
surements and history of exercise. The AST can also be falsely elevated by delay
in plasma separation, and release of AST from red cells.
In some instances the residual effects of glandular fever remain, or, the par-
ticipant has a high BMI and fatty liver. In either case the participant would not
be entered into the study.
Alkaline phosphatase is occasionally mildly elevated in the late, male
teenagers, reflecting possible bone injury in extreme sports or a late growth
spurt.
An unconjugated hyperbilirubinemia between 20 and 40 mol/L, and
occasionally up to 60 or 80 mol/L is a relatively common finding (up to 10%) in
the general population, and, in the absence of hemolysis and other hepatic disor-
ders, reflects Gilbert’s syndrome. Gilbert’s syndrome may become evident at any
age but is often seen in young adulthood associated with menstruation, fasting,
intercurrent illness, or dehydration. Fluctuating plasma bilirubin levels are typ-
ical, and the syndrome should not be interpreted as “disease.” Dubin–Johnson
and Rotor syndromes are likewise benign conditions that produce a conjugated
hyperbilirubinemia due to the failure of excretion of conjugated bilirubin.
An important adjunct to hepatic laboratory testing is, of course, the clinical
examination and history, as early cirrhosis may produce near normal or even
normal results for the above-mentioned tests.
Serological testing for hepatitis A, B, and C exclude participants with liver
damage from hepatitis B and C. Hepatitis A is diagnosed by the detection of IgM
anti-HAV (hepatitis A virus) during the acute illness, while IgG anti-HAV may
reflect a previous resolved episode of hepatitis A or vaccination.
A panel of antibody and antigen tests for hepatitis B and C will determine
the hepatitis status adequately in most instances, even in the early stages of dis-
ease. A note of caution is added, however, in participants who may have been
recently vaccinated, as we have found that vaccine antigens can be detected by
sensitive newer tests.
HIV testing must be performed with the knowledge that professional HIV
counseling is available should an unexpected positive test result occur, even
though HIV positive cases do not participate in a study. Counseling may also
be required for inconclusive first-line testing for other tests such as hepatitis B
and C, before confirmatory testing is available.
Renal function testing is an important part of screening to rule out renal
impairment that may affect drug excretion or drug handling by the body. Athletic
or body-building participants are particularly prone to the use of dietary supple-
ments such as proteins and creatinine, with consequent increases in plasma urea
and creatinine values. Conversely, significant renal impairment is possible while
plasma creatinine values remain within the reference range.
Adhering to conditions of testing is important. Glucose and cholesterol lev-
els need to be performed in the fasting state for reliable results. Furthermore, a
regular blood taking time, such as 8 AM, helps to negate circadian rhythm and
diurnal variation for some tests.
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 29
Repeat Examinations
While it is incumbent upon the clinical investigator to minimize participant dis-
comfort and time use, some tests and procedures need to be repeated to fully
determine their significance. These tests must be carefully followed and reported
to the participants (and/or family medical practitioner), especially if they have
been excluded from the study because of the initial abnormality.
Poststudy Testing
Laboratory tests are repeated at the end of the study. If abnormalities are detected
the test is usually repeated until it returns to normal (or nonsignificant) levels, or,
referred to the family medical practitioner for follow-up.
A Team Approach
Staff cognizant of the need for confidentiality initially greet and enroll partici-
pants and perform the social histories, ECG, blood pressure, height, and weight
recordings.
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
30 Hung et al.
The participant will meet several physicians during the reading, consent,
examination stages, and study periods. This provides the participant with a vari-
ety of opportunities for discussion, in both group and individual situations, and
with both physicians and nonphysician staff. In cases where doubt is cast on a
participant’s ability to understand the protocol or be willing to adhere to the
conditions of the study, a consensus of opinion is useful.
Study Design
Generally, if only two products are to be compared, a two-period, two-sequence
cross-over study design should be used. However, alternate, well-established
designs could also be considered such as a parallel design for substances with
a very long half-life (>100 hours).
For highly variable medicines (refer to Highly Variable Drugs and Drug
Product section), a replicate cross-over design should be considered. For exam-
ple, the following four-period, four-sequence cross-over design has been success-
fully employed for TGA registration of alendronate. In this study, the urine con-
centration of alendronate was measured and the bioavailability of alendronate,
as summarized by the accumulated excreted amount, was compared between
the two formulations.
Four-Period, Four-Sequence Cross-Over Design
Sequence Period 1 Period 2 Period 3 Period 4
1 A B B A
2 B A A B
3 A A B B
4 B B A A
Steady-State Studies
For most formulations, single-dose studies should generally suffice. However
there are instances where a steady-state design will be (i) required by the TGA
and Medsafe, for example, dose- or time-dependent pharmacokinetics and mod-
ified release products or (ii) considered, for example, where single dose admin-
istration does not allow adequate sensitivity for precise plasma concentration
determination or where it is intended to reduce intraindividual variability by
using a steady-state design. However, in some countries, steady-state studies are
no longer “considered” for the very reason that it dampens “true” variability
of a product as well as the ethical implications of multiple dosing to healthy
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 31
32 Hung et al.
Sample Size
The number of subjects should be determined statistically, however, generally
the minimum number of subjects starting the study should not be less than
12. The minimum number of completing subjects should be determined and
outlined clearly in the study protocol. In a cross-over design, the total variation
consists of sequence variation, subject (sequence) variation, period variation, for-
mulation variation, and error variation. For testing the formulation effect, the
error variation or intrasubject variation (18) is used. Therefore the sample size
required for a bioequivalence study should be estimated using the intrasubject
variation. In particular, since the logarithmic transformed AUC and Cmax val-
ues are used in constructing the 90% confidence intervals (CIs), the intrasubject
variation based on the lognormal distribution (19) must be used. However, this
intrasubject variation (or coefficient of variation, CV) can only be estimated if the
data of a previous bioequivalence study on the same formulation is available. In
most cases sample size is estimated from literature data. This typically consists of
only summary statistics such as average and standard deviation for the untrans-
formed AUC and Cmax values. Sample size estimation is highly dependent on the
standard deviation of the above pharmacokinetic parameters and the standard
deviation data provided in the literature do not usually provide information on
the intrasubject variation. In general, the CV calculated from published data on
standard deviation would be much larger than the intrasubject CV based on the
logarithmic transformed values. Therefore the sample size estimated from these
summary statistics is generally extremely conservative.
Data obtained from one of our studies on tramadol is used here to illus-
trate this discrepancy. In this study, the mean and standard deviation for AUC0–∞
(ng hr/mL) of the reference formulation were 2997.8 and 1259.2 respectively. The
CV estimated using only this information is 42%. With a CV of 42%, at a 5%
level of significance and 80% power, a minimum sample of 76 subjects would
be required even if the reference and the test formulation means were the same,
and a sample size of 94 would be required when the two means differed by 5%
(18). However, the mean squares error, obtained from the ANOVA table for the
logarithmic transformed AUC0–∞ values, was 0.03997637. By using the lognor-
mal distribution, the intrasubject CV was estimated to be 20% and a sample size
of 20 was large enough for a 5% difference in the two means (19). Twenty-four
subjects were included in this study and the resulting 90% CI for logarithmic
transformed AUC0–∞ was (0.975, 1.074). It is therefore obvious that a sample size
of 94 is excessive.
Washout Periods
Treatment periods should be separated by an adequate washout period. The
interval between study days should be long enough to ensure elimination of
the previous dose. The interval should be no less than five terminal elimination
half-lives of the active compound or metabolite. The interval between treatments
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 33
Sampling Schedules
The sampling schedule should provide an adequate estimation of Cmax and cover
the plasma concentration versus time curve long enough to provide a reliable
estimate of the extent of absorption. The Medsafe and TGA adopted guidance
indicates this is achieved if AUC0–t is at least 80% of AUC0–∞ . However, typically,
AUC0–t is at least 90% of AUC0–∞.
34 Hung et al.
Combination Products
Combination generic products should be compared with an equivalent innovator
combination product. However, in cases where there is no marketed innovator
product, separate products administered alone can be used for comparison. The
sample schedule should adequately provide for the determination of the phar-
macokinetic parameters of all active analytes and statistical analyses should be
conducted for all active analytes using the 90% CIs for all the active components
to fall within the usual acceptance ranges. However, in such cases, the combina-
tion product will be considered under a new medicine application (i.e., a new
formulation).
Over-the-Counter Products
The bioequivalence requirements for oral OTC products are the same as for pre-
scription only products However, both the TGA and Medsafe will consider a
bioequivalence study where the reference product is not sourced from Australia
or New Zealand provided that comparative dissolution testing and comparative
excipient analysis demonstrates that the reference products are essentially simi-
lar (refer to Test and Reference Products and Foreign Data section).
Australasia 35
subjects falls below the minimum. All subject samples should be analyzed, and
only those completing all phases of the study should be included in the final
pharmacokinetic and statistical analyses. Exclusion of entire subject data (out-
liers) is not generally accepted, except when a biological justification can be
proved, such as vomiting within twice the Tmax after dosing (23). Add-on stud-
ies as a rule are not permitted unless included in the original protocol. However,
revised analysis is allowed if the original statistical plan (e.g., AUC0–∞ ) proves to
be invalid. For example, if the results obtained in the study are markedly differ-
ent from the results referenced in published data or from previous studies, that is,
the sampling profile used in the study is not long enough to allow estimation of
AUC0–∞ or the accurate estimation of AUC0–∞ is difficult due to an erratic elimi-
nation phase and consequent difficulty in determining t1/2 , most likely the result
of endohepatic recycling. The original and revised results should be presented in
the final report (20).
Sample Analysis
All analytical methods used to determine the concentrations of the analyte(s)
and/or metabolites in the biological matrix must be fully validated and the
results documented (validation report). Validation procedures, methods, and
acceptance criteria must be specified in relevant standard operating procedures
(SOPs). A calibration curve should be constructed for each analyte in each ana-
lytical run and should be used to calculate the concentration of the analyte(s) in
the unknown (subject) samples. The calibration curve should consist of a blank
sample (drug-free plasma sample processed without internal standard), a zero
sample (drug-free plasma sample processed with internal standard), and an
appropriate number of nonzero plasma samples, including LLOQ (lower limit
of quantification) to cover the expected range of concentrations from the sub-
ject samples. Quality control samples [at three concentrations over the range of
the calibration curve, that is, one within 3 × LLOQ (low QC sample), one at the
geometric or arithmetic mean of the low and high QC sample (QC sample), and
one close to the upper boundary of the standard curve, that is, 80% of ULOQ,
(high QC sample)] should be included in the run in duplicate (six samples per
run). All the samples for each individual subject (all periods) should preferably
be medium analyzed in the same analytical run.
Analytes to be Measured
Parent Compound Versus Metabolite(s)
The analyte to be measured (23) in biological fluids collected in bioequivalence
studies is either the parent compound or when appropriate, its active metabo-
lite(s) (24). Measurement of only the parent compound rather than the metabo-
lite is generally recommended. The rationale for this recommendation is that the
concentration-time profile of the parent is more sensitive to changes in formula-
tion performance than a metabolite, which is more reflective of metabolite for-
mation, distribution, and elimination. However, the following are exceptions to
this general approach. Measurement of a metabolite may be preferred when the
concentrations of the parent compound are too low to allow reliable analytical
measurement in blood, plasma, or serum
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
36 Hung et al.
Australasia 37
Data Analysis
All pharmacokinetic analyses should be performed using validated com-
puter programs. Individual concentrations, AUC, Cmax , Tmax , and t1/2 calcu-
lations should be presented along with individual and mean plasma drug
concentration–time curves on both linear/linear and log/linear scales for each
completing subject. In addition, calculations for mean, geometric mean, standard
deviation, coefficient of variation, and ranges must also be presented. Spread-
sheets for each individual subjects’ raw data should be submitted. These should
include the following information: chromatogram identification, method file,
date and time of collection, retention times for the active compound(s) and inter-
nal standard(s), peak areas (heights) for the active(s) and internal standard(s);
calibration data, regression data and actual calculated drug concentrations. The
analytical report should also present a summary of data for each individual sub-
ject run, that is, mean and standard deviation of peak retention times, regres-
sion data, and “spiked” sample concentrations. Samples that require re-analysis,
according to a prespecified protocol, should be clearly identified and reasons
for re-analysis must be provided. The analytical report should also contain the
validation data. The method used to determine the active compound and/or
metabolites in a suitable matrix should be characterized, fully validated and doc-
umented (20). The main objective of the validation is to demonstrate the accept-
ability and reliability of the analytical results from the study and should include
the following information: (i) stability of stock solutions and of the analyte(s)
in the biological matrix under processing conditions and during the entire stor-
age period, (ii) specificity, (iii) accuracy, (iv) precision, (v) limit of quantification,
(vi) matrix effect when relevant, and (vii) dilution effect. The validation report
should include certificates of analysis for the analyte(s) and internal standard(s)
and representative chromatograms.
Calibration Curves
Calibration curves must be generated for each analyte in each analytical run and
used to calculate the concentration of the analyte in all subject samples. The stan-
dard curve fitting is determined by applying the simplest model that adequately
describes the concentration-response relationship by using appropriate weight-
ing and statistical tests for goodness of fit. Both x12 weighting and y12 weighting
are commonly used in regression calculations. Pateman (27) recommends a
weighting of x12 by assuming that “the SD of response tends to be proportional
to the concentration (x).” However, if no assumption is made on the SD except
that the CV is constant over the majority of the calibration range then the appro-
priate weight will be (␣+x)1
2 and the regression model is y = ␣ + x + ε . The
random error, ε , is unknown but the mean of the random error, E(ε) = 0 so it is
reasonable to replace ε by 0. This results in a weight of y12 , which is also called
the “empirical weight” (28). To compare the difference between the unweighted,
1
x2
weighting and y12 weighting a simple simulation can be performed by adding
10% to a single point in an ideal linear standard curve. This process is repeated
at each concentration and the absolute percentage deviation from the ideal con-
centration of the entire calibration curve standards is calculated. The results of
this simulation are presented in the table on page 38.
IHBK055-02
SPH
SPH
38
IHBK055-Kanfer
Standarda Unweightedb sum Abs.% deviation of 1/y 2 sum of abs.% Abs.% deviation of 1/x 2 sum of abs.% Abs.% deviation of
concentration of abs.% deviation the lowest and the deviation of all the lowest and the deviation of all the lowest and the
January 6, 2010
(g/mL) 10% added of all standards top standards c standards top standards c standards top standards c
1 9.97 (8.596,0.001) 11.04 (2.545,0.586) 11.08 (2.201,0.612)
2 12.75 (−2.802,0.002) 15.17 (-−3.066, −0.272) 16.04 (−3.582, −0.318)
4 18.27 (−5.575,0.004) 16.69 (−1.239, −0.653) 17.95 (−1.473, −0.777)
8 29.13 (−11.033,0.006) 16.68 (−0.358, −0.845) 17.93 (−0.425, −1.004)
16 50.16 (−21.598,0.010) 16.65 (0.082, −0.943) 17.88 (0.097, −1.118)
32 89.45 (−41.316,0.010) 16.94 (0.302, −0.992) 18.22 (0.357, −1.174)
18:19
64 156.94 (−75.070, −0.021) 17.08 (0.412, −1.017) 18.37 (0.488, −1.203)
128 247.30 (−119.658, −0.209) 17.15 (0.467, −1.029) 18.45 (0.552, −1.217)
256 248.90 (−116.960, −1.077) 17.18 (0.495, −1.036) 18.48 (0.585, −1.224)
Char Count=
460.8 260.77 (131.533, −3.722) 17.19 (0.507, −1.039) 18.50 (0.560, −1.227)
512 463.45 (236.620,4.898) 17.19 (0.509,8.857) 18.50 (0.601,8.649)
Mean 144.28 16.27 17.40
S.D 146.12 1.83 2.21
CV% 101.27 11.24 12.71
a Ten percent increase in the ideal peak area (height) ratios (PAR, range between 0.01 and 5.12 in this simulation) at one concentration each time while the ideal PARs of the
remaining concentrations are unchanged.
b Sum of the absolute% deviation from the estimated concentrations to the stated concentrations for all standards.
c Absolute% deviation of the lowest and highest standards.
Hung et al.
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 39
yi − ŷi 2
,
yi
Statistical Analysis
Pharmacokinetic parameters derived from the measures of concentration, for
example, AUC and Cmax should be analyzed using ANOVA. The data should
be transformed prior to the analysis by using a logarithmic transformation. The
statistical model includes the factors, sequence, subject (sequence) (i.e., subjects
nested within sequences), period, and formulation. The sequence effect should
be tested using the subject (sequence) mean square as the error term while the
other main effects should be compared to the mean square error obtained from
the ANOVA. Ninety percent CIs should be constructed for the ratio between
the test and reference formulation averages based on the logarithmic trans-
formed AUC and Cmax values using the Schuirmann’s two one-sided tests
procedure (29).
The pharmacokinetic parameters to be tested, the procedure for testing and
the acceptance criteria for each parameter should be outlined in the protocol. The
acceptance intervals for the primary pharmacokinetic parameters are as follows:
AUC (AUC0–t , AUC0–∞ , or AUC for steady-state designs)—the 90% CI
should lie within an acceptance interval of 0.80 to 1.25.
Cmax —the 90% CI should lie within an acceptance interval of 0.80 to 1.25.
As previously discussed, in certain cases a wider interval may be acceptable, but
must be justified and predefined in the protocol.
Tmax , t1/2 , and DF (Cmax −Cmin )/(AUC0–t/t ) for a steady-state design] are
considered to be secondary parameters and hence would not be used for evalu-
ating bioequivalence. However, 90% CI, based on nonparametric technique, for
untransformed data of Tmax and 90% CIs for the untransformed data of the other
secondary parameters are also presented for completeness.
Biowaivers
As previously indicated, an application for registration of a generic product
in either New Zealand or Australia should generally include a bioequivalence
study versus a leading brand (market leader) obtained from within the relevant
country. However, in certain well-defined circumstances, both authorities may
accept submission of foreign bioequivalence data, provided it can be shown that
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
40 Hung et al.
the local product and foreign product are identical, that is, supported by exten-
sive comparative dissolution testing and comparative excipient testing.
If an application pertains to several strengths of an active ingredient, a bioe-
quivalence study using only the highest strength may be acceptable. In this case
Medsafe and the TGA may grant a biowaver for the other strengths provided the
strength chosen for investigation has been justified and the following conditions
have been met:
1. The products are manufactured by the same manufacturer and using the
same process.
2. The active ingredient has demonstrated linear kinetics over the therapeutic
dose range.
3. The qualitative compositions of the different strengths are the same.
4. The ratio of the amounts of active ingredient and excipients is the same, or
where the concentration of the active ingredient is low (less than 5%), the
ratio of the amounts of excipients is similar.
5. The similarity should be justified by dissolution profiles covering at least
three time points, in three different buffers covering the physiological range,
that is, normally pH 1 to 6.8 (e.g., 1.0, 4.5, and 6.8), where the f 2 similarity
factor is >50 for the additional strengths and the strength of the batch used
in the bioequivalence study.
Australasia 41
estimate of the likelihood that the test product will be bioequivalent in the pivotal
equivalence study.
Safety Studies
When comparing topical corticosteroid formulations using a clinical end point
study, the safety of the product following application must be considered. For
example, one of the most obvious side effects of prolonged use of steroids is
hypothalamic–pituitary–adrenal (HPA) axis suppression. Typically this can be
addressed by using a parallel study design that monitors the plasma cortisol lev-
els before and after prolonged application (e.g., 4 weeks) of the test and reference
topical formulations. The area under the plasma cortisol concentration versus
time curve (AUC0–24 ) on Day 1 should be compared with the cortisol AUC0–24
on Day 29 to determine any significant difference between each treatment. The
test formulation will be deemed safe if the 90% CI is below the limit of 125%,
when compared to the reference formulation. If a toxicity study is required a ran-
domized, single topical application, irritancy study using synchronized applica-
tion and synchronized removal may be adopted. This design can be adopted to
show that there is no irritancy effect following the administration of a topical
formulation.
Nasal Sprays
At present, there is no guidance from the Authorities in either New Zealand or
Australia for demonstrating bioequivalence between nasal sprays for locally act-
ing products. However, protocols have been proposed by Sponsors to address
the efficacy and safety issues of nasal sprays for allergic rhinitis. The efficacy
study design is based on the draft guidance proposed by the FDA (33), which
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
42 Hung et al.
Inhalers
Both Medsafe and the TGA have developed guidance documents to demon-
strate bioequivalence of inhaled medications, Medsafe in Section 16 and TGA
in Appendix 19 of their respective guidelines. In general, the following protocols
address the requirements for exhibiting the bronchodilation and bronchoprotec-
tion effects of inhalers.
Bronchodilator Inhalers
Typically, therapeutic equivalence of bronchodilators, such as short-acting and
long-acting 2 -agonists, should be compared by their bronchodilatation potency
and efficacy to protect against bronchoconstriction caused by stimuli such
as methacholine (36), histamine (37), hypertonic saline (38), or exercise (39).
A double-blind, double-dummy, placebo-controlled, six-period, four-sequence
cross-over study, similar to that employed by Lavorini et al. (40) has been pro-
posed for the assessment of bioequivalence of salbutamol inhalers. Bioequiva-
lence is assessed by calculating the relative potency of two salbutamol formula-
tions by using either Finney’s 2 × 2 parallel regression analysis or the Emax model
(AUC0–60 of FEV1 vs. time). The relative potency is calculated and expressed as
the ratio of the estimated doses of salbutamol delivered by the two formulations
to achieve similar AUC0–60 values with Finney’s regression. If the Emax model
is used then the relative potency is estimated by the ratio of the ED50 values
obtained from the models. Two formulations are considered bioequivalent if the
90% CI of the relative potency is between 0.67 and 1.5. Whether this protocol can
be applied to long-acting 2− agonists, for example, salmeterol is not known. It
is our experience that the FEV1 is relatively insensitive to dose differences and
there is evidence that the commonly used doses of salmeterol may be higher
than required to produce a maximal effect. Other parameters such as PEFR (peak
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 43
expiratory flow rate) heart rate, QTc interval, plasma potassium concentration
and tremor in addition to bronchial reactivity can be measured to provide
unequivocal clinical effectiveness of the medicinal product (41).
Steroid Aerosols
The following protocols have been proposed for the evaluation of the bioequiva-
lence of inhaled steroids.
Cross-Over Designs
A randomized, double-blind, double-dummy, cross-over design with a 2- to 3-
week run-in period followed by a 4-week treatment period has been used. Each
treatment consists of either the test product and placebo as reference, or the ref-
erence product and the placebo as test. PD20 (36), after methacholine challenge
testing, is measured at the start and at the end of each treatment period.
1. Start of treatment period 1 (PD20 S1)
2. End of treatment period 1 (PD20 E1)
3. Start of treatment period 2 (PD20 S2)
4. End of treatment period 2 (PD20 E2)
For each subject, the difference of PD20 within each treatment is calculated:
D1 = PD20 E1 − PD20 S1 and D2 = PD20 E2 − PD20 S2
An ANOVA is performed on the logarithmic (base 2) transformed differ-
ences log2 (D) and a 95% CI is calculated on the log2 -scale. The formulations are
considered to be bioequivalent if the 95% CI is within the range (−1, 1).
Parallel Design
A randomized, double-blind, double-dummy, parallel group design with a 2-
week run-in period followed by a 6-week treatment period can be performed.
PD20 after methacholine challenge testing is measured at the start and at:
1. Start of run-in period (PD20 R)
2. End of run-in period (i.e., Start of treatment) (PD20 S)
3. After 3 weeks of treatment (PD20 M)
4. End of treatment period (PD20 E)
For each subject, the following difference will be calculated:
D = PD20 M − PD20 S or D = PD20 E − PD20 S
Analysis of covariance (with PD20 S as the covariate) is performed on the
untransformed differences D and a 90% CI is calculated on the untransformed
scale. The formulations are considered bioequivalent if both values of the 90% CI
are 0.8 or above (i.e., noninferior).
Side effects such as HPA axis suppression by inhalers can be determined
by measuring serum cortisol levels in healthy volunteers as described for topical
studies (35).
44 Hung et al.
Antiallergic Aerosols
Antiallergic aerosols such as sodium cromoglycate and nedocromil sodium are
generally less potent bronchodilators than 2− agonists and less potent bron-
choprotectors than steroids. Common side effects are limited to throat irrita-
tion on inhalation and unpleasant taste. A combination of the above-mentioned
study protocols may be useful for bioequivalence evaluation of the antiallergic
aerosols. However, these medicines are not effective in every subject and so the
demonstration of their effectiveness, on the provoked response, is a prerequisite
for entry into such studies (42).
REFERENCES
1. McBride WG. Thalidomide and congenital abnormalities. Lancet 1961; 2:1358.
2. Therapeutic Goods Act 1989—Australian Government Publishing Service, Canberra.
3. Medicines Act 1981 (Reprint 2006) (published under the Authority of the New
Zealand Government).
4. Australian Regulatory Guidelines for Prescription Medicines, June 2004. http://
www.tga.gov.au.
5. Module 1—Administrative Information and Prescribing Information for Australia.
Notice to Applicants CTD-Module, 1 September 2007.
6. Guidance on the GMP Clearance of overseas medicines manufacturers, 16th ed.
March 2008. http://www.tga.gov.au/manuf/gmpsom.htm.
7. New Zealand Regulatory Guidelines for Medicines, Vol 1, 5th ed. 2001. http://
www.medsafe.govt.nz.
8. Australian Regulatory Guidelines for OTC Medicines, July 2003. http://www.tga.
gov.au.
9. The Nuremberg Code from Trials of War Criminals before the Nuremburg Military
Tribunals under Control Council Law No. 10. Nuremberg, October 1946-April 1949.
Washington, D.C.: U.S. G.P.O, 1949–1953.
10. World Medical Association Declaration of Helsinki-Ethical Principals for Medical
Research involving Human Subjects 1964 (current version 2000).
11. Cartwright Inquiry. 2008. http://www.womens-health.org.nz/cartwright/cartwright
.htm.
12. Gisborne Cervical Screening Inquiry. 2008. http://www.csi.org.nz.
13. The Health Research Council of New Zealand, HRC Guidelines for Ethics Committee
Accreditation, June 2008.
14. New Zealand Researched Medicines Industry Guidelines on Clinical Trials Compen-
sation for Injury Resulting From Participation in an Industry Sponsored Clinical Trial.
2008. http://www.rmianz.co.nz.
15. Medicines Australia Form of Indemnity for Clinical Trials. 2008. http://www.
medicinesaustralia.com.au.
16. Jones B, Kenward MG. Design and Analysis of Cross-Over Trials. London: Chapman
and Hall, 1989.
17. Haider SH, Davit B, Chen ML, et al. Bioequivalence approaches for highly variable
drugs and drug products. Pharm Res 2008; 25:237–241.
18. Chow SC, Liu JP. Design and Analysis of Bioavailability and Bioequivalence studies,
2nd ed. New York: Marcel Dekker, 2000.
19. Hauschke D, Steinijans VW, Diletti E, et al. Sample size determination for bioequiv-
alence assessment using a multiplicative model. J Pharmacokin Biopharm 1992; 20:
557–561.
20. Note for Guidance on the investigation of bioavailability and bioequivalence
(CPMP/EWP/QWP/1401/98). CPMP Guidance-As adopted by the TGA-with
amendment. 10 April 2002. http://www.tga.gov.
21. Blume HH, Midha KK. Bio-International 92, Conference on bioavailability, bioequiv-
alence and pharmacokinetic studies. J Pharm Sci 1993; 2:1186–1189.
SPH SPH
IHBK055-02 IHBK055-Kanfer January 6, 2010 18:19 Char Count=
Australasia 45
3 Brazil
Margareth R. C. Marques
Department of Standards Development, U.S. Pharmacopeia, Rockville,
Maryland, U.S.A.
Sı́lvia Storpirtis
College of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
INTRODUCTION
The publication of the National Policy for Drug Products in 1998, the creation of
the National Agency for Sanitary Surveillance or Agencia Nacional de Vigilancia
Sanitaria (ANVISA), the approval of the law, and the publication of the tech-
nical guidances for the registration of generic products dramatically changed
the pharmaceutical market in Brazil. New concepts such as pharmaceutical
equivalency, therapeutic equivalency, bioavailability, and bioequivalence were
introduced (1–4).
The law and guidances for the registration of generic products in Brazil
were developed based on the regulations from countries or regions, such as
Canada, United States of America and the European Union, with a great deal
of experience with these type of products.
Brazil was the first country in South America to implement the evaluation
of pharmaceutical equivalency and bioequivalence studies for the registration of
generic products and nowadays is considered a model for the other countries in
the region.
A “similar” drug product is a product that contains the same drug(s) in the
same concentration, dosage form, route of administration, strength, and thera-
peutic indication as the reference drug product registered at the federal agency
in charge of sanitary surveillance, being allowed to differ only in characteristics
related to size and shape of the dosage form, expiry date, packaging, labeling,
excipients, and vehicles. It is always identified by its trade (branded) name and
not by the generic name.
Using the regulations for generic products (3,4) as a model, in 2003 new reg-
ulations and guidances were published for the registration of new similar prod-
ucts and for similar products already in the market. These regulations included
the evaluation of pharmaceutical equivalency, relative bioavailability, and good
manufacturing practices (GMP).
Similar products registered from May 2003 up to date must be in accor-
dance with the same regulations as generic products with the following
exceptions:
46
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
Brazil 47
Brazilian Regulations
The use of the generic name of the drug substance(s), according to the Brazilian
Common Nomenclature, together with the brand name on the packaging mate-
rial of products marketed in Brazil has been mandatory since 1980 (5,6).
The Law 793/93, published on April 5, 1993, defines the information on
the packaging materials of generic products. The generic name should be writ-
ten using a font with a size three times bigger than the brand name. The main
objective of this law was to stimulate competition on the drug product market
with subsequent reduction of product price. Unfortunately, the text of the law
was not very clear, with the main problem being the lack of demonstration of
therapeutic equivalency between generic and reference products (7).
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
48 Marques et al.
On May 14, 1996, Law 9279 establishing the regulations regarding intellec-
tual property was approved. This law was a significant step for the introduction
of new pharmaceutical dosage forms in the Brazilian market because there were
no regulations in Brazil for intellectual property rights for medicines and drug
products, allowing the registration of drug products, based only on its similar-
ity. This law was the first step toward regulations for the registration of generic
products similar to other countries with established and more modern regulatory
systems (8).
The regulations for generic drug products are part of the National Policy of
Drug Products, with the registration of this type of product being made accord-
ing to the following:
r Definition of criteria to demonstrate therapeutic equivalency, mainly regard-
ing bioavailability.
r Training and infrastructure of local laboratories to perform bioequivalence
studies.
r Incentive to produce generic drug products, laws and guidances for the
marketing, prescription and dispensing of generic products in the Brazilian
market (1).
On February 10, 1999, a new approved Law, 9787 (3), defined the con-
cept of a generic drug product and established the conditions for the use of the
generic drug name for all products. According to this law, the responsibilities of
ANVISA are
r to establish criteria and conditions for the registration and quality assurance
of generic products,
r to establish criteria for bioavailability studies of any drug product,
r to establish criteria for the verification of therapeutic equivalency through
bioequivalence studies to allow their interchangeability,
r to establish criteria for the dispensing of generic products. The decision to
replace the reference or branded product with a generic version is at the dis-
cretion of the practitioner (9). Items 3 and 5 of this law promote the prescrip-
tion and acquisition of generic drug products by all clinics and dispensaries
in the network maintained by the federal government, government actions
to facilitate the registration, sales and marketing of these products and facil-
itation of the information and education of the public and funding of special
programs to improve the quality of drug products. As a consequence of Law
9787/99 (3), a group of Brazilian experts in the areas of quality control, phar-
macology, and pharmaceutics was created to write the technical guidances for
the registration of generic products in Brazil. All the texts were revised by a
consultant with a great deal of experience on bioequivalence studies from the
University of Texas. As a result of the work of this team, on August 9, 1999,
Resolution RDC 391 (4) was approved. This document defined the technical
parameters for the registration of generic drug products and had the follow-
ing six annexures:
1) Guidance for stability studies.
2) Guidance for protocol and technical reports, bioequivalence, or bioavail-
ability studies.
3) Guidance for the validation of analytical methods.
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
Brazil 49
To implement all these new guidances and regulations and to review the
new dossiers submitted for the registration of this new category of products,
a new department within ANVISA, the General Management of Generic Prod-
ucts (Gerência Geral de Medicamentos Genéricos, GGMED) was created in
September 2000. The major actions undertaken by this new department were the
following:
Besides all these activities, ANVISA promoted and funded the installation
of Pharmaceutical Equivalency Centers and Bioequivalence Centers in the coun-
try. The former were accredited by ANVISA
In 2000, ANVISA introduced the Chamber of Regulations for Medicines
Market (Câmara de Regulação do Mercado de Medicamentos, CMED) to be
responsible for the management of the prices of generic drug products, main-
taining them at least 35% below the price of the reference product (12).
Since the introduction of generic products in Brazil, their sales have been
increasing at a very fast pace, and currently they represent about 16% of
medicines sold in Brazil (13).
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
50 Marques et al.
Bioavailability
ANVISA defines bioavailability as the rate and the extension of absorption of
a drug product in a dosage form, based on its concentration/time curve in the
systemic circulation or its excretion in urine (14).
Pharmaceutical Equivalency
According to ANVISA, pharmaceutical equivalents are drug products that con-
tain the same drug substance, in the same salt form or free base, in the same
amount, in the same type of dosage form, with or without the same excipi-
ents. These products should comply with the corresponding monograph in the
Brazilian Pharmacopeia. In the absence of a specific monograph in the Brazilian
Pharmacopeia, they should comply with the monograph in any other compendia
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
Brazil 51
accepted by the Brazilian health authorities, or with any other applicable qual-
ity standard. These quality standards include identity, strength, purity, potency,
uniformity, disintegration, and dissolution, when applicable. In Brazil, the eval-
uation of pharmaceutical equivalency should be done by a center qualified by
ANVISA (18). In June 2001, ANVISA implemented its quality system and posted
all standard operating procedures (SOPs) and criteria for the certification of phar-
maceutical equivalency centers on the agency website (www.anvisa.gov.br).
TABLE 1 Major Characteristics of the First Regulation for Generic Drugs Products in Brazil
Law Characteristics
√
√ Mandatory presubmission
RDC 391 √ Production and Quality Control Technical Report
(Aug 9, 1999) √ Manufacturing report for three pilot batches
Technical √ Validation of the manufacturing process
Guidance for the √ Name(s) of the supplier of the drug substance (not more than three)
Registration of √ Description of the synthesis route, including isomers and polymorphs
Generic √ Specifications and validated analytical methods
Products in √ Pharmaceutical equivalency
Brazil √ Certificate of GMPs and QC
√ Stability studies according to Zone 4
Bioequivalence—Quantification of the drug substance or metabolite
- Washout period of minimum of five t 1/2
- Number of healthy volunteers (in general 24)
- Weight variance ± 10% of the normal value
-√ Use of the Brazilian or international reference product.
Prescription: In the network by Brazilian Common Nomenclature
√ (DCB)—In private practice: brand name or DCB
Dispensing: Possibility of the pharmacist replacing the brand name
product by the generic version, if there are no restrictions by the
private practitioner
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
52 Marques et al.
Brazil 53
54 Marques et al.
Study Design
An open, cross-over, randomized design is required where the volunteers receive
test and reference drugs on separate occasions (periods), in a single- or multiple-
dose regimen. If necessary, a parallel design can be used, for long terminal half-
life drugs (longer than 24 hours).
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
Brazil 55
Subjects
Number of Subjects
The number of subjects included must provide adequate statistical power for
bioequivalence demonstration. Numbers of participants can be calculated from
the coefficient of variation (intraindividual) and statistical power, and no less
than 12 volunteers may be used. If no literature data are available the investigator
can opt for the inclusion of at least 24 volunteers. The guidance for planning the
statistical issues of bioavailability/bioequivalence studies [Resolution 898 from
2003 (24)] describes a method to calculate the number of volunteers based on the
procedure described by Chow and Liu (25).
Selection of Subjects
Sex/Age
Subjects must be 18 years of age or older and capable of giving informed consent.
They may be of either/both sexes but if males and females are included, there
must be an equal distribution among the study sequences. If the drug product
is to be used in a specific population (age and gender) the investigator must
include volunteers of this population. As an example, bioequivalence studies of
oral contraceptives must include women of child-bearing age.
Mass
The weight of the subjects should be within 15% of the normal range for males
and females, taking into account their height and physical type. It also recom-
mends the inclusion of woman within 10% of the normal range in bioequivalence
studies for oral contraceptives.
Informed Consent
Informed consent must be approved by an ethics committee that has to be sub-
mitted for ANVISA evaluation together with the bioequivalence study report.
The informed consent form must contain the drug name, the dose by unit, the
pharmaceutical form, and the name of the manufacturer(s). Detailed information
about the informed consent content is presented in Resolution 196 from 1996 (26).
Medical Screening
The Guidance for bioavailability/bioequivalence studies [Resolution 1170 from
2006 (23)] does not state any information regarding medical screening but the
Guidance for protocol of bioavailability/bioequivalence studies [Resolution 894
from 2003 (27)] states that the protocol must contain the clinical evaluation of the
volunteers, a list of the laboratory tests (blood, biochemistry, hepatitis B and C,
HIV, beta-HCG for women, type I urine tests, and electrocardiogram).
56 Marques et al.
Phenotyping/Genotyping
The Brazilian regulations for bioavailability/bioequivalence studies do not dis-
cuss this topic.
Brazil 57
and depot dosage forms, the duration and the frequency of sampling should
be sufficient to characterize the absorption, distribution, and elimination of the
drug. For long terminal half-life drugs (longer than 24 hours), an alternative
collection time, for at least 72 hours, or a parallel design may be used.
Number of Samples
The Brazilian regulations for bioavailability/bioequivalence studies do not dis-
cuss this topic.
Characteristics to Be Investigated
Blood/Plasma/Serum Concentration Versus Time Profiles
The pharmacokinetic parameters should be calculated based on plasma drug
concentration versus time profile. When dealing with endogenous compounds,
the baseline level should be measured and the analysis must be done with and
without basal correction. The following pharmacokinetic parameters should be
presented: AUC0–t , AUC0–∞ , Cmax , tmax , and the half-life, t1 . For multiple-dose
/2
studies, the parameters should be AUC0– , Cmax , tmax , Cmin , C avg , and the fluctu-
ation index where
AUC0–t —Area under the plasma/serum/blood concentration–time curve from
time zero to time t, where t is the last time point with measurable concen-
tration for individual formulation;
AUC0–∞ —Area under the plasma/serum/blood concentration–time curve from
time zero to time infinity.
tmax —Time to maximum plasma concentration.
Cmax —The maximum observed plasma concentration after dose administration.
Pharmacodynamic Studies
Pharmacodynamic studies are indicated when it is not possible to measure the
drug in plasma or in urine in an accurate way, for example, ophthalmic suspen-
sions and local acting inhalers.
Bioanalysis
Details about performing and validating bioassays are stated in the Guidance for
bioanalytical method validation [Resolution 899 from 2003 (28)].
58 Marques et al.
Validation
Specificity
Analyses of blank samples of the appropriate biological matrix (blood, plasma,
serum, urine, or other matrix) should be obtained from at least six sources (four
normal, one lipemic, and one hemolyzed). Each blank sample should be tested
for interference, and selectivity should be ensured at the lower limit of quantifi-
cation (LLOQ).
Precision and Accuracy
Precision should be measured using a minimum of five determinations per con-
centration (high, middle, and low). The precision determined at each concentra-
tion level should not exceed 15% of the coefficient of variation (CV) except for
the LLOQ, where it should not exceed 20% of the CV. Accuracy is determined by
replicate analysis of samples containing known amounts of the analyte. Accuracy
should be measured using a minimum of five determinations per concentration
(high, medium, and low). The mean value should be within 15% of the actual
value except at LLOQ, where it should not deviate by more than 20%.
Lower Limit of Quantification
The limit of quantification should be at least five times the response compared
to blank response or analyte peak (response) should be identifiable, discrete, and
reproducible with a precision of 20% and accuracy of 80% to 120%.
Limit of Detection
The limit of detection (LOD) should be at least two or three times the response
compared to blank response.
Recovery
The recovery of the analyte does not need to be 100%, but the extent of recovery
of an analyte and of the internal standard should be consistent, precise, and exact.
Recovery experiments should be performed by comparing the analytical results
for extracted samples with unextracted standards that represent 100% recovery.
Response Function
The simplest model that adequately describes the concentration–response rela-
tionship should be used. Selection of weighting and use of a complex regression
equation should be justified.
Robustness
This parameter should be considered during the method development phase. If
measurements are susceptible to variations in analytical conditions, the analyti-
cal conditions should be suitably controlled or a precautionary statement should
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
Brazil 59
Calibration Curves
A calibration curve should be generated for each analyte in the sample. A suf-
ficient number of standards should be used to adequately define the relation-
ship between concentration and response. Concentrations of standards should
be chosen on the basis of the concentration range expected in a particular study.
A calibration curve should consist of a blank sample (matrix sample processed
without internal standard), a zero sample (matrix sample processed with internal
standard), and six to eight nonzero samples covering the expected range, includ-
ing LLOQ. The following conditions should be met in developing a calibration
curve: 20% deviation of the LLOQ from nominal concentration and 15% devi-
ation of standards other than LLOQ from nominal concentration. At least four
out of six nonzero standards should meet the above criteria, including the LLOQ
and the calibration standard at the highest concentration. The exclusion of any
standards should not change the model used.
60 Marques et al.
Reassay
Chromatographic analyses
Reintegration
Others.
Data Analysis
The statistical evaluation should be done according to the Guidance for planning
and accomplishment of the statistical phase of bioavailability/bioequivalence
studies [Resolution 898 from 2003 (24)].
Statistical Analysis
Pharmaceutical parameters shall be obtained from the blood concentration
curves of the drug versus the time, and statistically analyzed to determine the
bioequivalence; in case of endogenous substances, statistical analysis should
be conducted using quantified plasma concentrations with and without base-
line level correction, while the decision on bioequivalence should be based on
the corrected values.
The following pharmacokinetic parameters should be determined:
r AUC0–t , AUC0–∞ , Cmax of the drug and/or metabolite, and the time to reach
this peak (Tmax ) should be directly obtained, without data interpolation; elim-
ination half-life (t1/2 ) of the drug and/or metabolite should also be deter-
mined, although there is no need for statistical treatment.
r For evaluation of bioequivalence, the parameters AUC0–t , Cmax , and Tmax
should be used and in the case of multiple-dose studies, steady state after
administration of the test and reference drugs must be confirmed.
r The exclusion of more than 5% of volunteers that participated in the study is
not permitted until the conclusion or absence of over 10% of the blood concen-
tration values of the drug from the administration of each drug per volunteer.
Data Presentation
a) Present a table containing individual values, averages (arithmetical and geo-
metrical), standard deviation, and variation coefficient of all pharmacokinetic
parameters related to the administration of test and reference drugs.
b) It is recommended that the AUC0–t and Cmax parameters be transformed into
natural logarithms.
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
Brazil 61
Single-Dose Studies
The following pharmacokinetic parameters have to be presented in the report:
Cmax , tmax , AUC0–t , AUC0-∞ , and t1 . The 90% CI for Cmax and AUC0–t has to be
/2
between 80% and 125%.
Reporting of Results
The Resolution 895 from 2003 (29) describes all the information that should be
presented in the bioequivalence report, which includes the test and reference
data, administrative information, responsible personnel and sites, study proto-
col, ethics committee approval, and the specific information about each phase.
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
62 Marques et al.
Clinical Report
The documents should include study design, randomization list, drug informa-
tion, drug accountability, and all the information of the study population (demo-
graphic, laboratory tests), inclusion and exclusion criteria, time of drug adminis-
tration and blood collections, fasted and fed times, dose washout period, adverse
effects, and protocol deviations.
Analytical Report
Should contain bioanalytical method description and validation, reference stan-
dards, method to calculate drug concentrations, complete information about the
analytical runs (date, initial and final time, volunteers analyzed, calibration stan-
dard and bias, quality controls, and bias), reanalysis (initial and reanalysis val-
ues, cause for reanalysis, criteria for choosing the value), and complete series of
chromatograms from 20% of the volunteers.
Quality Assurance
The Resolution does not say anything about quality assurance, but the study can
be performed only in contract research organizations (CROs) previously certified
by ANVISA.
Suspensions
A bioequivalence study is always required.
Brazil 63
64 Marques et al.
Tablets—Lower Strength
Tablets with different strengths/doses, in the same pharmaceutical dosage form
and with proportional formulations, same release mechanism, manufactured
by the same producer, at the same site, a bioequivalence study should be per-
formed with the higher dosage strength and a bioequivalence study for the other
strengths can be waived if the dissolution profiles are comparable among all
strengths according to the Guidance for assay dissolution for solid oral phar-
maceutical forms [Resolution 310 from 2004 (32)]. For modified release dosage
forms, a comparative dissolution study using three different dissolution media
(such as pH 1.2; pH 4.5, and pH 6.8 media) should be performed. In addition, the
dissolution profile should be comparable among test and reference dosage forms
for all dosage strengths.
REFERENCES
1. Brazil. Health Minister. National Health Surveillance Agency. MS/GM Ordinace no.
3916 of October 30, 1998. Approves the National Drug Policy. Brası́lia, DF; 1998.
http://e-legis.bvs/leisref/public/showAct.php?id=751. Accessed June 16, 2009.
2. Brazil. Law no. 9782 of January 26, 1999. Defines the National Health Surveillance
System, establishing the National Health Surveillance Agency, among other pro-
visions. Brası́lia, DF; 1999. http://e-legis.bvs/leisref/public/showAct.php?id=182.
Accessed June 16, 2009.
3. Brazil. Law no. 9787 of February 10, 1999. Amending the Act no 6360, to Septem-
ber 23, 1976, which deals with health surveillance, provides the generic product,
provides for the use of generic names in pharmaceutical and other provisions.
SPH SPH
IHBK055-03 IHBK055-Kanfer December 28, 2009 11:36 Char Count=
Brazil 65
66 Marques et al.
for the manufacture and quality inspection of drugs. Brası́lia, DF; 1995. http://
e-legis.bvs/leisref/public/showAct.php?id=5355. Accessed June 16, 2009.
20. Brazil. Health Minister. National Health Surveillance Agency. RDC Resolution no.
210 of August 4, 2003. Determines all establishments manufacturers of drugs, com-
pliance with the guidelines established in the technical regulation of the prac-
tice for the manufacture of drugs. Brası́lia, DF; 2003. http://e-legis.anvisa.gov.br/
leisref/public/showAct.php?id = 22321&word = Accessed June 16, 2009.
21. Brazil. Health Minister. National Health Surveillance Agency. RDC Resolution no.
134 of May 29, 2003. Available on the suitability of drugs already registered Brası́lia,
DF; 2003. http://e-legis.bvs/leisref/public/showAct.php?id=7904. Accessed June
16, 2009.
22. Brazil. Health Minister. National Health Surveillance Agency. RDC Resolution no. 136
of May 29, 2003. Provides for the registration of new drug. Brası́lia, DF; 2003. http://e-
legis.bvs/leisref/public/showAct.php?id=7914. Accessed June 16, 2009.
23. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no.
1170 of April 19, 2006. Determines the publication of guidelines for evidence
on bioavailability/bioequivalence of drugs. Brası́lia, DF; 2006. http://e-legis.bvs/
leisref/public/showAct.php?id=21746&word. Accessed June 16, 2009.
24. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no. 898
of May 29, 2003. Guide to planning and performing the step of statistical studies on
the bioavailability/bioequivalence. Brası́lia, DF; 2003. http://e-legis.anvisa.gov.br/
leisref/public/showAct.php?id = 2489&word = Accessed June 16, 2009.
25. Chow SC, Liu, JP. Design and Analysis of Bioavailability and Bioequivalence Studies.
New York: Marcel Dekker, 2000.
26. Health Minister. National Health Surveillance Agency CNS Resolution no. 196
of 10 October 10, 1996. Requirements for conduct of clinical research using
health products for humans. Brası́lia, DF; 1996. http://e-legis.anvisa.gov.br/leisref/
public/showAct.php. Accessed June 16, 2009.
27. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no.
894 of May 29, 2003. Determines the protocol for publication of the guide and
technical report of the bioequivalence study. Brası́lia, DF; 2003. http://e-legis.bvs/
leisref/public/showAct.php?id=1914. Accessed June 16, 2009.
28. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no.
899 of May 29, 2003. Determines the publication of guidelines for validation of
analytical and bioanalytical methods, is repealed RE Resolution no 475 of March
19, 2002. Brası́lia, DF; 2003. http://e-legis.bvs/leisref/public/showAct.php?id=5745.
Accessed June 16, 2009.
29. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no.
895 of May 29, 2003. Determines the publication of “Guidelines for preparation of
technical report on the study of bioavailability/bioequivalence”. Brası́lia, DF; 2003.
Accessed June 16, 2009.
30. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no. 897
of May 29, 2003. Determines the publication of guidelines for relief and replace-
ment of bioequivalence studies. Brası́lia, DF; 2003. http://e-legis.bvs/leisref/public/
showAct.php?id=1775. Accessed June 16, 2009.
31. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no. 893
of May 29, 2003. Determines the publication of the guide to make changes, addi-
tions and reporting post-registration of drugs. Brası́lia, DF; 2003. http://e-legis.bvs/
leisref/public/showAct.php?id=1909. Accessed June 16, 2009.
32. Brazil. Health Minister. National Health Surveillance Agency. RE Resolution no. 310
of September 1, 2004. Determines the publication of the guide for the study and
reporting of pharmaceutical equivalence and profile of dissolution. Brası́lia, DF; 2004.
http://e-legis.bvs/leisref/public/showAct.php?id=12431. Accessed June 16, 2009.
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
4 Canada
Iain J. McGilveray
McGilveray Pharmacon Inc., Ottawa, Ontario, Canada
INTRODUCTION
As a result of 1969 amendments to the Patent Act (1) (compulsory licensing),
Canadian regulators were the first to apply pharmacokinetics (PKs) to safety and
efficacy risk assessment of generic drug products. However, formal guidelines
developed by an Expert Advisory Committee (EAC), currently referred to as the
Scientific Advisory Committee (SAC), were not published until the 1990s. Cur-
rently, guidelines are being updated and new SAC initiatives such as Guidances
for nonproportional PK drugs and for drug products requiring fed studies have
been published in draft form for stakeholder comments and consultation while
others, such as guidance for critical dose drugs, have been finalized.
The Canadian Food and Drugs Act and consolidated Regulations in Divi-
sion 8 (2) require that for new drugs (which also means drug products), the man-
ufacturer must file a New Drug Submission (NDS) that must provide evidence of
safety, efficacy, and consistency of quality. (This requirement includes all drugs
submitted after 1962, but for some older drugs, it may be applied if presented for
new claims or in a new dosage form or strength.) However, for second and sub-
sequent entry new drugs (generic drug products), an abbreviated NDS (ANDS),
the regulation was revised in 1995 as C.08.002.1., noting that a manufacturer may
file an abbreviated new drug submission (ANDS) for the generic product (i.e.,
“the new drug”), where, in comparison with a Canadian reference product,
a. the new drug is the pharmaceutical equivalent of the Canadian reference
product;
b. the new drug is bioequivalent with the Canadian reference product, based on
the pharmaceutical and, where the Minister considers it necessary, bioavail-
ability characteristics;
c. the route of administration of the new drug is the same as that of the
Canadian reference product;
d. the conditions of use for the new drug fall within the conditions of use for
the Canadian reference product.
In this regulation “pharmaceutical equivalent” means a drug product that,
in comparison with another drug product, contains identical amounts of the
identical medicinal ingredients, in comparable dosage forms, but does not nec-
essarily contain the same nonmedicinal ingredients; and “Canadian Reference
Product C.08.001.1.” (http://www.canlii.org/ca/regu/crc870/secc.08.001.1.html)
means:
a. a drug in respect of which a notice of compliance is issued pursuant to Section
C.08.004 and which is marketed in Canada by the innovator of the drug,
67
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
68 McGilveray
b. a drug, acceptable to the minister, that can be used for the purpose of demon-
strating bioequivalence on the basis of pharmaceutical and, where applica-
ble, bioavailability characteristics, where a drug in respect of which a notice
of compliance has been issued pursuant to Section C.08.004 cannot be used
for that purpose because it is no longer marketed in Canada, or
c. a drug, acceptable to the minister, which can be used for the purpose of
demonstrating bioequivalence on the basis of pharmaceutical and, where
applicable, bioavailability characteristics, in comparison to a drug referred
to in paragraph a
In general, this means an innovator product, but there is a policy (3) for
exceptions when the innovator has withdrawn from the Canadian market. This
regulation was developed after application of internal policies and decisions by
reviewers of applications over about 30 years. These ad hoc policies resulted after
amendments to the Canadian Patent Act in June 1969 (1), which made provision
for compulsory licensing and facilitated registration of generic drug products in
Canada. [“In a compulsory license, a government forces the holder of a patent,
copyright or other exclusive right to grant use to the state or others. Usually, the
holder does receive some royalties, either set by law or determined through some
form of arbitration” (4). In Canada, this royalty was set at 4%.]
The Patent Act amendment in Canada, facilitated growth of a substantial
generic market (the first in a developed country), although the amendment was
attenuated in 1988 when the patent life was extended to international norms. Fol-
lowing the legal changes, introduction of generic products led to a need for assur-
ing the safety and efficacy of such products and a research program into compar-
ative bioavailability was undertaken in the 1970s. In fact, Health Canada was
the first jurisdiction to apply bioequivalence (BE) to safety and efficacy review of
new drug products (5).
Many of the internal decisions for generic product approval involved an
Expert Advisory Committee on Bioavailability first formed in 1971, which was
at that time largely a review committee for early BE studies carried out in Health
Canada laboratories. However, the EAC not only provided decisions from review
of study results for generic studies, but also was the first to define bioequiv-
alence from comparative bioavailability and to provide an early standard that
such products should have a bioavailability of at least 80% relative to a “refer-
ence formulation” according to a statistically sound design (6).
The major work of the EAC toward the end of the 1980s was the produc-
tion of three reports A, B, and C that were the basis of two guidelines (7,8) and
several policies that provide the foundation of most of this chapter. There is
also draft guidance, “Preparation of Comparative Bioavailability Information for
Drug Submissions in the CTD Format” (9), which provides a list of the require-
ments to be met and integrates unique Canadian bioequivalence requirements
with the ICH harmonized approach (Common Technical document, CTD) for
drug product registration, to which Canada is a signatory.
However, it must be understood that Canada is governed as a Confedera-
tion of Provinces. Thus, the regulations and guidelines for BE are federal, lead-
ing to a Notice of Compliance (NOC) to sponsors for marketing in Canada and
while their application leads to a Declaration of Bioequivalence for specific prod-
ucts, the Provinces in the Canadian Confederation have the responsibility for
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
Canada 69
health care and the licensing of health professions. Thus, provincial formulary
committees consider interchangeability of products and may not accept the fed-
eral decision of BE (which also evaluates quality) to be sufficient to list a particu-
lar product. Also, each province has different formulary rules, usually permitting
product substitution of prescribed brand name by pharmacists and also govern-
ing prices. Of course, before being approved federally with a Notice of Com-
pliance, the generic product must comply with the patent laws. This has been
problematic in some cases, as slight changes in innovator products have gained
extensions and often delayed introduction of generic products into the market.
Most of the Canadian requirements for bioavailability and particularly BE
are, as previously mentioned, provided in two guidances, Part A for immediate-
release or conventional oral products (7) and Part B for modified-release (MR)
products (8). Both the guidances, Part A and B, state that “this guidance docu-
ment deals with drugs that have uncomplicated Characteristics.”
Part A provides examples of drugs that may require methodology and stan-
dards to be modified for certain drugs (products)—for example, those with one
or more of the following characteristics:
a. MR dosage forms.
b. Complicated or variable PKs, for example,
r nonlinear kinetics;
r substantial first-pass effect (greater than 40%);
r variable kinetics owing to different genetic phenotypes;
r stereochemical effects such as in vivo inversion of configuration;
r an effective half-life of more than 24 hours.
c. (An important) time of onset of effect or rate of absorption.
d. High toxicity or a narrow therapeutic range.
e. Little or no absorption with activity exerted locally in the gastrointestinal
tract.
f. A drug measurement methodology insufficiently sensitive or reliable to
determine blood concentrations to at least three terminal half-lives.
g. Combination products.
h. Biologicals.
Guidances for these types of drugs were never finalized, but report C of
the EAC (10) suggested modifications for some of the above and, more recently,
some specific guidances or policies have been developed for these exceptions, as
will be discussed later in this chapter.
Study Design
This is described under Section 4 of the guidance, “Study Design and Environ-
ment,” which is introduced with the statement “The design of a bioavailability
study should minimize variability that is not attributable to the drug per se and
should eliminate bias as much as possible. The guidances in this section serve for
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
70 McGilveray
the usual case. Other designs may be permissible after consultation with Health Canada
(HC) before the study is initiated.”
Section 4.3 states that “The basic design to be used is a two-period cross-
over, in which each subject is given the test and reference formulations at differ-
ent times in a blinded manner). In cases where more than two formulations are
under study, or are studied under different conditions, each volunteer should
receive all treatments in a restricted randomized design. However, when the
number of treatments results in a study that is longer than a month, a balanced
incomplete block design may be considered.” The latter type of design is rarely
used.
Canada 71
Section 3.2.c deals with health checks and since adoption of the ICH good
clinical practice (GCP) guidance (11) the Health Canada guidance has been
superseded, although in general the statement “The health of the volunteers
must be determined by the supervising physician through a medical examina-
tion and review of results of routine tests of liver, kidney, and hematological
functions” covers many of the GCP expectations. Psychological evaluation of
candidate subjects is also required to exclude patients unlikely to comply with
study restrictions or unlikely to complete the study. Also, when the drug has a
cardiac effect, an electrocardiogram should be included in the study documen-
tation. Of course, as in the GCP guidance (11), full documentation of aberrant
laboratory values should be rechecked and a summary must be presented along
with the physician’s opinion. While there is no actual direct mention of informed
consent from volunteers, the requirement for ethical review board approval (now
according to clinical trial certification (12) with advice for BE applications (13)
and citation of good clinical practice guidelines (now ICH) are very clear on doc-
umentation of consent, as well as responsibilities of investigators and suitability
of clinical, laboratory and analytical facilities involved in the study. For details
see also “CTD preparation” document (13).
Nonsmoking subjects are preferred, but if smokers are included they must
be identified, with discussion on any likely effect on the study included in the
study report. Volunteers should not take any other drug, including alcoholic bev-
erages and over-the-counter (OTC) drugs, for an appropriate interval before, as
well as during, the study (Section 4.1). In the event of emergency, the use of any
drug must be reported (dose and time of administration). It is expected that alco-
hol and other recreational drug abuse would be detected during health checks.
There is no direct mention of genotyping or phenotyping in the older guidance.
However for subject safety, GCP requires that poor metabolizers be protected
from adverse effects.
72 McGilveray
Canada 73
Characteristics To Be Investigated
Blood/Plasma/Serum Drug Concentration Versus Time Profiles
Section 4.8 of the guidance A discusses sampling of blood or urine. “Under nor-
mal circumstances, blood should be the biological fluid sampled to measure the
concentrations of the drug. In most cases the drug may be measured in serum or
plasma; however, in some cases, whole blood may be more appropriate for anal-
ysis. If the concentrations in blood are too minute to be detected and a substantial
amount (>40 percent) of the drug is eliminated unchanged in the urine, then the
urine may serve as the biological fluid to be sampled. When urine is collected,
the volume of each sample must be measured immediately after collection and
included in the report. Urine should be collected over no less than three times
the terminal elimination half-life. For a 24-hour study, sampling times of 0 to 2,
2 to 4, 4 to 8, 8 to 12, and 12 to 24 hours are usually appropriate. Quantitative
creatinine determinations on each urine sample are also required. Sometimes the
concentration of drug in a fluid other than blood or urine may correlate better
with effect. Nevertheless, the drug must first be absorbed prior to distribution to
the other fluids such as the cerebrospinal fluid, bronchial secretions, and so on.
Thus, for bioavailability estimations, blood is still to be sampled and assayed.”
Examples of drugs for which urinary excretion profiles may be used for
BE are potassium chloride and biphosphonates. Pharmacodynamic studies are
rarely used for BE, but report C has a section on this (10). It is a very general
advice. Of interest is the suggestion on appropriate standards:
The requirements of a pharmacodynamic study should be comparable to
those of standard bioavailability or bioequivalence studies, including mea-
sures of the magnitude, onset and duration of response. For approval under
such circumstances, criteria similar to those defined for bioavailability and
bioequivalence studies that use drug concentration measurements must
be derived; e.g., AUC of measured pharmacodynamic response and maxi-
mum response. In addition, similar standards should be met in these crite-
ria to establish bioavailability and bioequivalence.
In fact such studies are drug specific and usually wider standards are nec-
essary. The human skin blanching assay (HSBA) for topical corticosteroid prod-
ucts and the change in pulmonary function tests with albuterol are the known
applications and are noted later.
Bioanalysis
Section 6 of the guideline, Part A, entitled “Measurement Methodology”
describes the attributes of such methods for parent drug, and when appropriate,
metabolites in the matrix and the validation procedures required in reports to
establish and maintain selectivity, range, precision, and accuracy. There is a draft
guidance on metabolites (15), which essentially notes that parent drug should be
measured for BE, even for prodrugs, except if the sensitivity does not permit the
performance standards to be met to obtain an adequate profile. The 1992 guid-
ance refers to the report (16) of a bioanalytical methods validation workshop
(the first of its kind), which laid out internationally accepted practices for this
area but has been superseded by the report of a subsequent (second) conference
(17). The report describes the requirements for method development validation
and then partial validations required for changes in processes, methodology, or
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
74 McGilveray
analytical equipment, for example, different mass spectrometers with the final
validation being the application to the study samples. Documentation requires
listing of standard operation procedures (SOPs) for sample storage, processing,
standard and quality control sample preparation, and evaluation of stability (for
analyte and internal standard) under various conditions, such as benchtop as
well as long-term frozen samples. A Canadian difference compared to interna-
tional practice requirements, given in the Part A guidance, was that there was to
be reanalysis of 15% randomized replicate (or incurred) samples for each study,
but this was revoked in 2005 (18). However, the during the May 2006 third bioan-
alytical methods validation conference discussed the need for validating meth-
ods by using incurred as well as the spiked samples normally evaluated for pre-
cision and accuracy. The “15% randomized replicate” request was an attempt
to demonstrate reproducibility in practice. However, it was difficult to apply as
other jurisdictions did not require incurred sample stability in that era. In fact
determination of the need (or not) for incurred sample investigation may be
expected in the original bioanalytical method validation report rather than the
report for each study (Eric Ormsby, TPD, personal communication, May 2008).
Canada 75
Data Analysis
Section 7 of guideline, Part A details the PK and statistical analysis expected.
Statistical Analysis
The requirements for “analysis of data” are described in Section 7.0 of the guid-
ance. The analysis of variance (ANOVA) tables submitted with the study doc-
umentation (report) should include the appropriate statistical tests of all effects
in the model. The output from ANOVAs appropriate to the study design and
execution must be expressed with enough significant figures to permit further
calculations.
The analyses should include all data for all subjects on measured data. Supple-
mentary analyses may also be carried out with selected points or subjects initially
excluded from the analyses. Such exclusions must be justified. It is rarely accept-
able to exclude more than 5 percent of the subjects or more than 10 percent of the
data for a single subject-formulation combination.
In fact, rejection of outliers is rarely accepted and remains an issue to be resolved,
despite several advisory committee discussions. In the impending revision of
Canadian guidelines it will be proposed (20) that values from subjects identified
as extreme with an acceptable outlier test can be rejected, if after retesting the
subjects with both formulations, the retested values are found to be within the
normally expected range.
An ANOVA should be carried out on the time to peak concentration, tmax
and terminal slope () data, and on the logarithmically transformed AUCT ,
AUCI , and Cmax data. The analysis and results for each parameter should be
reported on a separate page as detailed in the guidance (see Section 8, “Sample
Analysis for a Comparative Bioavailability Study”). The reported results must
include
a. means and CVs (across subjects) for each product;
b. the ANOVA, containing source, degrees of freedom, sum of squares, mean
square, F and p values and the derived intra- and intersubject CVs;
c. AUCT and Cmax ratios for test versus reference products;
d. the 90% confidence interval (CI) about the mean AUCT ;
e. estimates of measured content for each formulation being compared, and a
separate table showing points c and d above, corrected for measured content.
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
76 McGilveray
Add-On Studies
There is an allowance for add-on studies. The latter thus permits two or more
studies to be pooled if certain requirements are met, viz
a. the same protocol must be used for all studies. Specifically, this means that
the same analytical method is to be used, the blood samples drawn at the
same time, and the same lots of the same formulations used.
b. two consistency tests must be done on the studies to ensure that pooling is
meaningful. The first test is the test of equality of the residual mean squares
and the second test is the formulation by study interaction.
Canada 77
78 McGilveray
reference product, calculated for each study subject. To date this standard
has only been applied to two drugs, oral gel formulations of ibuprofen, and
tablet formulations of sumatriptan.
Another update of report C was the Notice to Industry: Bioequivalence
Requirements for Combination Drug Products (24). The purpose is to state
BE requirements specific to combination drug products. Further work is being
done on requirements for fixed dose combinations, particularly in New Drug
Submissions.
For studies of combinations, the PK parameters to be reported and assessed
according to acceptance criteria, are those which would normally be required of
each drug if it were in a formulation as a single entity, as described in current
TPD guidelines (Parts A and B) and policy statements (which are given earlier).
There is a draft policy on Bioequivalence Requirements: Drugs Exhibiting
Non-Linear Pharmacokinetics (25), which states that a drug will be considered to
exhibit nonlinear PK if this is indicated in the peer-reviewed scientific literature
or the approved labeling for the drug. However, the drug may be treated in the
same way as those exhibiting linear PKs, if evidence is provided to show that
dose-normalized AUC deviate (increase or decrease) by less than 25% over the
labeled dose range for the proposed indication.
Drugs that exhibit nonlinear PK characteristics with single or multiple
doses of approved strengths should meet standards for BE as outlined in the TPD
Guideline on the Conduct and Analysis of Bioavailability and Bioequivalence Studies:
Part A or Part B, as applicable.
These requirements should be met in single-dose studies in both the fasted
and fed states for all nonlinear drugs, with the following exceptions:
a. If nonlinearity occurs after the drug enters the systemic circulation unless
there is evidence that a product exhibits a food effect;
b. If a condition (fasted or fed) for product ingestion is contraindicated, that
condition may be waived in a BE trial.
This draft policy is also under consideration (20) and it is proposed that
if the product monograph for the reference clearly states the type of nonlinear-
ity, then the drug is considered to be nonlinear (non–dose-proportional) and the
appropriate dose should be used in the study or studies. A fed study would not
be required.
Steady-State Studies
The major group of drugs with different criteria are MR products as described
in the guideline, Part B (9). This guidance defines the general acceptance crite-
ria for the required multiple-dose, steady-state studies, which are also applica-
ble to multiple-dose studies that may be required for some nonlinear drugs and
with the narrower limits previously described, for critical dose drugs. However,
the application of steady-state studies to extended-release products is also being
reconsidered (20) in harmony with the FDA general guidance. Steady-state stud-
ies do not seem to provide any better comparison of the two formulations. The
general acceptance criteria for BE from steady-state studies are described in Sec-
tion 4.3 of the guideline, Part B, and are based on the geometric mean ratio of
test to reference and the standards described below must be met for parameters
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
Canada 79
calculated from the observed concentrations, as well as those corrected for mea-
sured drug content.
AUC: The 90% CI of the relative mean AUC (generally known as AUCss : AUC
at steady state for the dosage interval) of the test to reference formulation
should be within 80% to 125%.
Cmax : The relative mean measured Cmax at steady state of the test to reference
formulation should be within 80% to 125%.
Cmin : The relative mean measured Cmin at steady state of the test to reference
formulation should not be less than 80%.
Reporting of Results
As Health Canada applies the ICH guidances and the CTD to submission review,
submissions are required to follow the subsections of this document. There is an
additional advice in the draft guidance for industry “Preparation of Comparative
Bioavailability Information for Drug Submissions” in the CTD Format, Health
Canada, May 2004 (9). Thus the clinical report, analytical report, and PK and
statistical reports would be included in CTD Module 5 Section 5.3.1, Biopharma-
ceutic studies, with reports according to the ICH good clinical practice guideline,
including “adverse events.” In addition, the Part A and other TPD guidance stan-
dards and tabulations would be expected. Quality information must be included
in the quality overall summary and details in CTD Module 2, Quality.
Solutions
There is no specific guidance for PK studies of solutions, including bioavailabil-
ity of solutions of new drugs. However, during development, basic PK studies
in support of general safety and efficacy would be expected. To integrate with
ICH guidelines, there is a relatively new guidance on Pharmaceutical Quality
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
80 McGilveray
Suspensions
Bioequivalence studies are expected for oral suspensions for systemic effects
according to the 1995 Drugs Directorate Guidelines, Preparation of drug identi-
fication number submissions (27). Demonstration of bioequivalence is generally
required and oral suspensions, powders/granules for oral suspension, intended
for systemic effect containing prescription drugs are affected. The standards for
AUCT and Cmax given in the guideline, Part A (7), are identical to those listed
below for immediate-release conventional formulations of uncomplicated drugs.
For complicated drugs, such as critical dose drugs, the acceptance ranges are as
described in “Data Analysis” section of this chapter.
Canada 81
82 McGilveray
For group II, first market-entry MR dosage forms, the same single- and
multiple-dose PK studies as already described for group I products are required.
The studies should be generally pursued in the context of demonstrating the
efficacy and safety of the recently developed drug product, and comparable
bioavailability should be demonstrated against an appropriate reference formu-
lation. The single-dose study should be a comparison between a single dose of
the first market-entry MR formulation and the doses of the innovator’s conven-
tional formulation that the MR formulation is intended to replace. (The doses
of the conventional formulation are administered according to the conventional
dosing regimen.) When identical doses of conventional and MR formulations
cannot be administered, a proportionality correction must be made for the cal-
culation of relevant parameters. Unless there are subject safety concerns, both
fasted and fed studies should be completed. In addition for group II products,
steady-state studies are required for formulations used at a dose interval likely to
lead to accumulation (AUCX /AUCI < 0.8 for the MR product). The comparison
should be made between the first market-entry MR formulation and equivalent
doses of the conventional formulation over the dosing interval (claimed for the
MR product) at steady state. Generally, steady-state studies should be performed
under fasting conditions unless subject safety might be compromised, in which
situation a fed study may be applied. In this case, manufacturers should consult
with Health Canada before undertaking a study.
The bioavailability standards below are suggested for group II products. It
should be noted that usually these types of products are supported by extensive
clinical trials and such information can justify results of comparative bioavail-
ability versus immediate-release reference that do not meet these standards (usu-
ally lower). The fed study is both an attempt to check for dose-dumping and to
provide labeling information.
Acceptance criteria suggested for group II products:
r AUC—The geometric mean ratio AUC of the MR formulation to the conven-
tional formulation should be between 80% and 125% in the fasting state.
The AUC may be evaluated by determining AUCT , provided that AUCT
obtained by the linear trapezoidal rule is at least 80% of the extrapolated AUCI
(i.e., AUCT /AUCI ≥ 0.80).
r Cmax —The geometric mean ratio of Cmax of a single dose of the MR formu-
lation to the conventional formulation should not exceed 125% in the fasting
state.
In some cases, the intended use of the MR formulation may call for a mod-
ification of the stated Cmax criterion. In such cases, before undertaking the
study, manufacturers should consult with Health Canada.
Canada 83
a AUC may be evaluated by determining AUCT , provided that AUCT obtained by the linear
trapezoidal rule is at least 80% of the extrapolated AUCI (i.e., AUCT /AUCI ≥ 0.80).
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
84 McGilveray
Canada 85
While the policy is intended for topical NSAIDs, the principles outlined
therein with respect to the safety issue of sensitization could be applied
to any topically administered product that has shown potential for
sensitization.
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
86 McGilveray
Canada 87
88 McGilveray
Canada 89
when a product is marketed in more than one strength, if the formulation of each
strength contains the same ingredients in the same proportion (i.e. the formula-
tions are proportional), the results of a single comparative bioavailability study
can be extrapolated to all strengths in the series. Extrapolation becomes more dif-
ficult, however, when the proportion of ingredients changes among the strengths
or when there are pre- or post-marketing formulation changes. In general if dif-
ferent strengths are proportional in formulation, or have only ‘minor’ differences
in the proportion of ingredients, a comparative bioavailability study is required
for only one strength (preferably the highest).” Differences in proportion are con-
sidered to be “minor” when no strength within a range differs from a studied
strength, by more than the percentages listed in the policy for various classes
of excipients (e.g., <5% for filler, <0.25 for magnesium stearate as lubricant).
The total additive effect of all excipient changes should not be more than 5%.
Changes in coatings that are not designed to play a role in the drug release mech-
anism are also generally concluded to be “minor.” In all instances, if compara-
tive bioavailability data are not provided for each formulation, the sponsor must
provide scientific justification for a waiver of this requirement. This justification
will include dissolution using a validated QC method. In this regard, a validated
method is one that has been demonstrated to be sensitive to changes in formula-
tion and manufacturing, including the physicochemical attributes of formulation
ingredients, as documented in method development studies. In the absence of a
validated method, the comparative dissolution profiles should be determined in
three media, as described in “Choice of Reference Product—C.08.001.1.” section.
Media should be selected to emphasize possible differences between the prod-
ucts, for example, a medium in which the dissolution rate is relatively slow (e.g.,
pH of the medium close to the pKa value of the drug) may offer some advan-
tages. The percentage of drug content released should be measured at a number
of suitably spaced time points, for example, at 10, 20, and 30 minutes, and con-
tinued to achieve virtually complete dissolution. At least six dosage units of each
should be tested.
The policy applies only to drugs with uncomplicated characteristics as
defined in the guideline, Part A. Comparative bioavailability studies would
likely be required for all strengths of critical dose drugs for generic (or formula-
tion change) approval [see, Guideline Part A, Section 5.1 and may be considered
on a case-by-case basis, Ref. (36).]
90 McGilveray
with the BE study of higher strength allowing a waiver of an in vivo study for
lower strengths, if the appropriate quality attributes, including dissolution pro-
files are documented and appropriate.
CONCLUSIONS
While the general Canadian guidelines for BE have many similarities to the FDA
and European guidances, there remain differences that can cause difficulties in
submitting second entry products (generics) to TPD. The Canadian reference or
comparator situation in general still requires studies to be repeated if a foreign
reference has been used in an ANDS (3). This is in distinction to an NDS when
the company can provide full details of global strategy. The nonlinear draft guid-
ance (25) is different from other jurisdictions and often results in an additional
fed study being required for Canadian approval. The correction for potency is
still required (but note below). As yet, for biowaivers, the Canadian regulators
have not embraced the BCS, or at least made clear the tolerances for acceptance
using the approach. Clearly for oral dosage forms for systemic use, the guide-
lines are clear and comprehensive. Nonoral, locally acting drugs (such as dermal
or inhaled routes), with few exceptions require clinical trials. The exceptions are
when some relevant pharmacodynamic measures can be applied, such as with
corticosteroids [dermal (30) and inhaled (32)].
As has been noted, within the Health Canada, Therapeutic Products
Directorate, changes to the general guideline Ab are under consideration (20),
although, the draft for consideration has not been released at the time of writing
this chapter ( June 2009). The objective is a revision of the guideline Part A (7)
to remove ambiguities. It will also include Guideline Part B (8) for MR products
and attempt to integrate report C classes (10) as well as modifications of other
special guidancesc or policies on BE.
With the caveat that these are not yet final, the main points for change are
provided below:
b Guidance 1 will combine current “Conduct and analysis” guidelines Parts A (1992) and B
(1996)—clarifies some positions and adds some new approaches. Guidance 2—defines classes
of drugs and their required studies and standards (Report C drugs, 1994).
c Guidance 3—data requirements and standards for inhaled corticosteroid products used in
the treatment of asthma. Use of steroid-naive patients with mild to moderate asthma, and
3% eosinophil count and FEV1>60%. Three-treatment (reference, test, and placebo) parallel
study design for three weeks by using the lowest dose of the Canadian Reference Product and
eosinophil recommended as primary outcome measure (also FeNO, FEV1). Secondary mea-
surements (FEV1, asthma symptoms) where both test and reference must be 50% over placebo
(% change from baseline). Test versus Reference 90% CI between 80% and 125% and systemic
absorption test to reference must meet usual BE standards. Guidance 4—data requirements
and standards for nasal steroid products.
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
Canada 91
d For “add-ons”—consideration being given to use a Bonferroni adjustment to keep error rate
at 5% and remove consistency tests.
e Sequential design being considered when variation and expected mean differences are uncer-
tain, viz, expected CV and mean difference to be stated, total N determined from table, decla-
ration of n < N where interim analysis will be performed; 93% CI to be applied as penalty for
stopping early and BE accepted. N.B. All Must Be Defined in Study Protocol.
f Adaptive designs, similar to sequential design being considered to allow re-estimation of sam-
ple size based on observed CV of the new study, viz, estimation of expected CV and mean
difference and determination of sample size N; n < N to be defined where interim analysis
will be performed; reestimation of sample size required based on observed CV. N.B. Must Be
All Defined in Study Protocol.
g More flexibility to be provided in analysis using mixed effects methodology (e.g., Proc mixed
in SAS) since mixed effects models analyze all types of cross-over designs (replicate) and han-
dles missing values, that is, when subject misses one period; per-protocol analysis not intent-
to-treat analysis being considered.
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
92 McGilveray
still give well-characterised profiles i.e., not caused by poor placement of sam-
pling times.” Currently (7) (but very rarely) 5% of subjects can be removed
with justification. The proposals include application of nonparametric 90%
CIs or allowing removal of identified extreme values from an acceptable out-
lier test after retesting the subject(s) identified with both test and reference
formulations. The reanalysis with new values would indicate if the outliers
can be removed. If the subject continues to show up as an outlier, there is a
problem.h
r Incurred sample analysis will not be required for BE studies, but in bioanalyt-
ical method development the “reproducibility of samples must be shown to
be adequate for the intended purpose.”
Some other areas to be considered are the following:
r Highly variable drugs and drug products (HVDP)i
r Definition of an “acceptable plasma profile”
r Dealing with endogenous substances, for example, adjusting for baseline
r How best to analyze urinary excretion data when concentrations in blood or
its components are too low to be quantified for BE assessment.
REFERENCES
1. Bill C-102 An Act to Amend the Patent Act, the Trademarks Act and the Food and
Drugs Act. http://www. curc.clc-ctc.ca/lang. Accessed September 2008.
2. Department of Justice Canada, http://laws.justice.gc.ca/en/F-27/C.R.C.-c.870/
234078.html. Current to February 21, 2006.
3. Health Canada, Drugs and Health Products Drugs Directorate Policy regarding
the use of a non-Canadian Reference Product under the provisions of Section
C.08.002.1(c) of the Food and Drug Regulations. December 1995. http://www.
hc-sc.gc.ca/dhp-mps/prodpharma/applic-demande/pol/crp prc pol-eng.php.
Accessed September 2008.
4. http://en.wikipedia.org/wiki/Compulsory license. Accessed September 2008.
5. McGilveray IJ. Bioequivalence: A canadian regulatory perspective. In: Welling PG, Tse
FLS, Dighe SV, eds. Pharmaceutical Bioequivalence. Drugs and the Pharmaceutical
Sciences, Vol 48. New York: Marcel Dekker, Inc., 1991:381–418.
6. Morrison AB, Cook D, Casselman WGB. Clinical Equivalency: A health protection
branch perspective. Can Med Assoc J 1973; 109:800–808.
h Consideration being given to define an outlier as a value which is 3 standard deviations away
from mean; does not overlap with other observations; has a studentized residual greater than
at the 0.02 level. Two approaches to deal with outlier, viz, construction of a nonparametric
90% CI or two allowance of removal of identified subjects’ data after retesting subjects with
both formulations and new results not identified as outliers using same approach. These con-
siderations are based on the premise that cause of extreme values unlikely to be formulation
related.
i HVDPs defined as drugs where intrasubject CV of AUC and/or Cmax greater than 30%. Rec-
ommendations being considered, viz: outlier analysis to be done; adjustment of bioequiva-
lence interval (BI) as a function of the intrasubject CV in a stepwise manner as CV increases,
for example, if observed CV from study is between 30% and 39% BI will be 77% to 130% and
likewise if observed CV from study is between 60% and 69% BI will be 61 to 160%, which will
allow sample sizes to be significantly reduced while retaining 80% power and means at 100%.
Intention to provide consistent decision-making for same Canadian Reference Product from
different sponsors.
SPH SPH
IHBK055-04 IHBK055-Kanfer December 28, 2009 11:39 Char Count=
Canada 93
94 McGilveray
22. Guidance for industry, Bioequivalence Requirements: Critical Dose Drugs, Health
Canada, May 2006. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-
demande/guide-ld/bio/critical dose critique-eng.php. Accessed September 2008.
23. Notice to industry. Bioequivalence Requirements for Drugs for Which an Early
Time of Onset or Rapid Rate of Absorption Is Important (rapid onset drugs).
Health Canada, June 2005. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-
demande/guide-ld/bio/notice rapidonset avis apparitionrapide-eng.php. Accessed
September 2008.
24. Notice to industry: Bioequivalence Requirements for Combination Drug Products,
Health Canada, June 2005. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-
demande/guide-ld/bio/notice avis comb-eng.php. Accessed September 2008.
25. Draft policy on Bioequivalence Requirements: Drugs Exhibiting Non-Linear
Pharmacokinetics, Health Canada, July 2003. http://www.hc-sc.gc.ca/dhp-mps/
prodpharma/applic-demande/pol/nonlin pol-eng.php. Accessed September 2008.
26. Guidance for industry, Pharmaceutical Quality of Aqueous Solutions. Health
Canada, February 2005. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-
demande/guide-ld/chem/aqueous aqueuses-eng.php. Accessed September 2008.
27. Drugs Directorate Guidelines, Preparation of drug identification number sub-
missions, Health Canada, 1995. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/
applic-demande/guide-ld/din/pre din ind-eng.php. Accessed September 2008.
28. Policy issue “Submissions for generic parenteral drugs”, Health Canada, 1990.
http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-demande/pol/gen subm
pres pol-eng.php. Accessed September 2008.
29. Policy issue Submissions for generic topical drugs, Health Canada, 1990. http://
www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-demande/pol/gener pol-eng.php.
Accessed September 2008.
30. Policy on Submissions for Topical Non-Steroidal Anti-inflammatory Drugs (Top-
ical NSAID’s). http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-demande/
pol/topnsaids ainstop pol-eng.php. Accessed September 2008.
31. Guidance to establish Equivalence or Relative Potency of Safety and Efficacy
of a Second Entry Short-Acting Beta2-Agonist Metered Dose Inhaler (MDI).
http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-demande/guide-ld/inhal-
aerosol/mdi bad-eng.php. Accessed September 2008.
32. Draft Guidance Document: Submission Requirements for Subsequent Market
Entry Inhaled Corticosteroid Products for Use in the Treatment of Asthma, May
2007. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-demande/guide-ld/
inhal corticost-eng.php. Accessed September 2008.
33. Draft guidance document Submission Requirements for Subsequent Market Entry
Steroid Nasal Products for Use in the Treatment of AllergicRhinitis. http://www.hc-
sc.gc.ca/dhp-mps/prodpharma/applic-demande/guide-ld/nas rhin-eng.php. Ac-
cessed September 2008.
34. Health Canada Policy Issues From the Drugs Directorate Changes to marketed new
drug products, April 1994. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/applic-
demande/pol/changmar nd dn pol-eng.php. Accessed September 2008.
35. 5 Post-Notice of Compliance Changes: Quality Document, July 2006. http://www.
hc-sc.gc.ca/dhp-mps/prodpharma/activit/proj/post-noc-apres-ac/noc postnotice
ac apreavis-eng.php. Accessed September 2008.
36. Health Canada Policy issue from the drugs directorate Bioequivalence of Propor-
tional Formulations: Solid Oral Dosage Forms, March 1996. http://www.hc-sc.gc.ca/
dhp-mps/prodpharma/applic-demande/pol/bioprop pol-eng.php. Accessed Sep-
tember 2008.
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
Joelle Warlin
Federal Agency for Medicines and Health Products, Brussels, Belgium
INTRODUCTION
The generic drug product market is projected to grow from US $15 billion in
2004 to US $27 billion in 2009 in the United States, and from US $9 billion to
US $14 billion in Western Europe (1). Moreover, the growth opportunities for
generic drug products in the near future are significant with an estimated US
$100 billion worth of branded pharmaceutical products to go off patent by 2010
(1). The substantial growth of the world generics drug market has been driven
by a number of factors, but in particular the need to contain public health care
spending, including the expenditure on drug products. In response to the impor-
tant growth of the generic pharmaceutical industry during the last 10 to 15 years,
regulatory agencies in countries all over the world, such as the Food and Drug
Administration (FDA) in the United States, Canada’s Health Products and Food
Branch (HPFB), and the European Medicines Agency (EMEA) in the European
Union (EU), have established requirements which must be met by a generic drug
product to receive marketing authorization (2,3).
The EU offers four routes for the registration of generic drug products: (i)
a national procedure, (ii) a mutual recognition procedure (MRP), (iii) a decen-
tralized procedure (DCP), and (iv) a centralized procedure (CP) (4). The national
procedure may lead to marketing authorization of the generic drug product in
the concerned member state. This national procedure is still being used, but
is strictly limited to medicinal products that are not authorized in more than
one member state. The MRP is based on the principle of mutual recognition of
national authorizations and, therefore, provides for the extension of marketing
authorizations granted to one member state, the so-called reference member state
(RMS), to one or more member states identified by the applicant. Since Novem-
ber 2005, the applicant may make use of the DCP and submit an application to
each of the member states where it is intended to obtain a marketing authoriza-
tion and choose one of them as the RMS. The RMS prepares a draft assessment
report and collects all comments received from the concerned member states that
are forwarded to the applicant. Further steps are managed by the RMS to reach
a consensus and to finalize the procedure.
Since 2004, it is possible to apply for marketing authorization of medici-
nal products in the EU by using the CP. According to this procedure, a single
95
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
a Norway, Iceland and Liechtenstein form the European Economic Area (EEA) with the 25 mem-
ber states of the EU. These countries have, through the EEA agreement, adopted the complete
EU acquis on medicinal products and are consequently parties to the EU procedures. Where in
this text reference is made to member states of the EU this should be read to include Norway,
Iceland and Liechtenstein.
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
necessary, reference will be made to the new revised EMEA Guideline on the
Investigation of Bioequivalence but it should be kept in mind that, at this stage,
it is only a draft version (10).
Study Design
For many drugs a large intersubject variability in pharmacokinetic parameters,
such as the extent of absorption (F), the apparent volume of distribution (V),
and plasma clearance (CL), is generally observed. The intrasubject variability
usually is substantially smaller than the between-subject or intersubject variabil-
ity and, therefore, a cross-over design is generally recommended for BE stud-
ies (12,13). The EMEA Note for Guidance on the Investigation of Bioequiva-
lence and Bioavailability is clear in this regard and recommends a two-period,
two-sequence cross-over design, with random allocation of the subjects to each
sequence, when comparing the bioavailability of two medicinal products (7).
Other study designs may be acceptable, such as a parallel study design for
long half-life substances and replicate designs for substances with highly vari-
able pharmacokinetics (14). In the case of a cross-over design, treatments should
be separated by a sufficiently long washout period (usually at least five times the
terminal plasma half-life of the active drug substance or its metabolites) to ensure
that all of the drug and/or its metabolite(s) has been cleared from the body prior
to the time of the subsequent administration.
The number of subjects required for a BE study should ideally be estimated
at the design stage and is determined by (i) the error variance ( 2 ) of the primary
BE metrics to be studied, (ii) the significance level (␣), (iii) the expected deviation,
with respect to the primary BE metrics, between the two formulations which is
considered compatible with BE (e.g., ± 20% for AUC), and (iv) the required sta-
tistical power (15,16). An estimate of the error variance can be obtained from
the published literature, a previous BE study, or by undertaking a pilot study.
Nomograms of the number of subjects required for various ratios of the expected
means for test and reference products and various intrasubject coefficients of
variation have been published by Diletti et al. (15,17). The guidance document of
Canada’s HPFB allows an add-on study (stated a priori in the protocol) when the
results from the first study fail to reach the required power, under the condition
that appropriate statistical tests validate the analysis of the combined data (18).
The draft version of the revised EMEA Guideline on the Investigation of Bioe-
quivalence includes a recommendation regarding under which circumstances a
sequential design (a so-called two-stage approach) may be used (10).
For oral immediate release dosage forms the EMEA guidance favors a
study where a single dose is taken on an empty stomach, that is, following
an overnight fast, with a fixed volume of fluid (at least 150 mL). However, if
it is recommended in the summary of product characteristics (SPC) that the
reference medicinal product should be taken with a meal, the BE study should be
carried out under fed conditions if the recommendation of food intake has any
pharmacokinetic implications such as a higher bioavailability (9). All subsequent
meals and drinks as well as other test conditions (e.g., posture during the first
few hours following intake of the medicinal products, physical activity, etc.)
should be standardized to minimize the variability in the bioavailability metrics
unrelated to a possible difference in the formulations. For the same reason of
minimising variability, BE studies are recommended to be carried out in healthy
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
volunteers, of either sex, between 18 and 55 years old having a normal body
weight based on body mass index, preferably nonsmokers, and without a history
of alcohol or drug abuse. For an active substance known to be subject to major
genetic polymorphism in its metabolic elimination, phenotyping/genotyping
“should be considered” according to the EMEA Note for Guidance on the
Investigation of Bioavailability and Bioequivalence when using a parallel design
(7). Phenotyping/genotyping “may be considered” as well for crossover BE
studies for safety or pharmacokinetic reasons (7). Indeed, plasma concentra-
tions of an active substance that is a substrate for an enzyme showing genetic
polymorphism may be much higher and half-lives much longer in poor metab-
olizers, thus necessitating longer sampling schedules compared to extensive
metabolizers (19).
Although, in general, a single-dose study will suffice to show that a generic
drug product is bioequivalent to an approved reference product, according to the
EMEA Note for Guidance on the Investigation of Bioavailability and Bioequiv-
alence there are situations in which steady-state studies may be required or can
be considered (7). These situations may include BE studies for active substances
undergoing dose- or time-dependent kinetics, or for active substances with high
intraindividual variability for which it may be difficult or even impossible to
demonstrate BE in a reasonably sized single-dose study. In addition, steady-state
studies can be considered when problems of analytical sensitivity preclude suf-
ficiently precise measurement of analyte plasma concentrations after single-dose
administration. According to the revised EMEA Guideline on the Investigation
of Bioequivalence, a multiple-dose study as an alternative to a single-dose study
may also be acceptable if problems of sensitivity of the analytical method pre-
clude sufficiently precise plasma concentration measurements after single-dose
administration. However, if possible, Cmax should be determined as a measure
of peak exposure following administration of the first dose of the multiple-dose
study. AUC, a measure of extent of exposure, should be determined at steady
state. Moreover, in a multiple-dose BE study the administration scheme should
preferably follow the highest usual dosage recommendation (10).
Post hoc exclusion of outliers based on pharmacokinetic or statistical rea-
sons alone is not accepted (7,9). Nonstatistical reasons to exclude the data
of a particular subject from the final statistical analysis should have been
prospectively defined in the protocol, or according the EMEA Questions &
Answers document on the bioavailability and BE guideline: “. . . at the very
least, established before reviewing the data.” Acceptable explanations to exclude
pharmacokinetic data or to exclude a subject from the final statistical anal-
ysis would be protocol violations such as vomiting, diarrhoea, analytical
failure, etc.
Concerned Member States may request information from the first Mem-
ber State on the reference product, namely on the composition, manu-
facturing process and finished product specification. Where additional
bioequivalence studies are required, they should be carried out using
the product registered in the concerned Member State as the reference
product.
BE Metrics
The area under the plasma (serum, blood) concentration of the parent compound
versus time curve (AUCt , AUC∞ ) generally serves as a measure of the extent of
absorption. Tmax and the corresponding maximum plasma concentration, Cmax ,
may serve as characteristics of the rate of absorption. However, it should be
emphasized that Cmax is not a pure measure of absorption rate but is confounded
with the extent of absorption (23). Urine excretion data may also be used to
determine the extent of absorption provided elimination is predominantly renal
as intact drug substance and is dose proportional (7,9). In the revised EMEA
Guideline on the Investigation of Bioequivalence the condition that “elimination
is dose-linear and is predominantly renal as intact drug” is no longer mentioned
(10). However, the use of urinary data has to be carefully justified when used to
estimate the rate of absorption (9,10,24).
AUC∞ , the area under the curve extrapolated to infinity, can only be reli-
ably measured if the terminal plasma half-life can be accurately determined,
which is not always the case. AUCt , the area under the curve from the time of
administration to the last measurable plasma concentration at time t, is there-
fore considered to be the most reliable measure of the extent of absorption
provided that it covers at least 80% of AUC∞ . Literature data support the
notion that BE assessment for long half-life drugs is not adversely affected by
using truncated AUC (25–29). Blood sampling time in this case should be suf-
ficiently long to ensure completion of gastrointestinal transit of the drug prod-
uct (approximately two to three days) and consequently the absorption pro-
cess of the drug substance. The Canadian HPFB guidelines, for example, accept
AUC0–72 , the AUC from time 0 to 72 hours following administration, as a mea-
sure of extent of absorption for drug substances with a half-life of more than
12 hours (30).
The most recent version of the general BE guidance from the FDA requests
that only the parent compound should be measured to assess BE (34). Only when
a metabolite is formed as a result of gut wall or other presystemic metabolism
and the metabolite contributes to safety and efficacy is the metabolite measured
to provide supportive evidence. In all other instances only the parent compound
is measured for BE. According to the guidelines of Canada’s HPFB, the determi-
nation of BE is based on measurement of the active ingredient, or its metabolite,
or both, as a function of time (18). They further specify that normally measure-
ment of the parent compound is sufficient but in some cases measurement of
the metabolite could be required. For example, when a prodrug is administered,
the active metabolite should be measured. The Questions & Answers document
on the Bioavailability and Bioequivalence Guideline which were recently formu-
lated by the CHMP Efficacy Working Party of the EMEA, also deals with the
issue when metabolite data should be used to establish BE (9). This document
stipulates that metabolite data can only be used if the applicant presents state-
of-the-art evidence that measurements of plasma concentrations of the parent
compound are unreliable. In addition, it is pointed out that Cmax of the metabo-
lite is less sensitive to differences in the rate of absorption than Cmax of the parent
compound:
Therefore, when the rate of absorption is considered of clinical importance,
bioequivalence should, if possible, be determined for Cmax of the parent
compound if necessary following administration of a higher dose. (9)
active substance are interchangeable in any individual patient. For this reason,
the “two one-sided tests” procedure is based on the intrasubject variability
that is commonly estimated from the mean square error (MSE), also called the
residual mean square, of an analysis of variance in which the fixed effects are
typically formulation, period, sequence, and subject nested within sequence.
The intrasubject variability can be estimated from the MSE by calculating the
CVanova (ANOVA coefficient of variation) as follows:
CVanova (%) = e MSE − 1 100
The width of the 90% CI depends on the magnitude of MSE and the number
of subjects in the BE study. Active substances whose AUC and Cmax show a high
intrasubject variability have high values for CVanova (>30%) and are called highly
variable drugs (HVDs). The larger the CVanova , the higher the number of subjects
required to give adequate statistical power (16,17).
The usual acceptance limit for the 90% CI around the geometric mean ratio
for AUC and Cmax , that is, 0.80 to 1.25 (or 80–125%), is based on a consensus
amongst clinical experts that a difference of ±20% in plasma concentrations of
the active substance following administration of two different medicinal prod-
ucts would have no clinical significance for most drugs (36). Since measures
derived from plasma concentrations such as AUC and Cmax are log-normally
distributed, this ±20% translates into an asymmetric acceptance limit, for exam-
ple, 0.80 to 1.25. The EMEA Note for Guidance suggests that in specific cases of
active substances with a narrow therapeutic index (NTI) the acceptance interval
may need to be tightened but does not give more specific information (7). The
draft version of the EMEA revised Guideline on the Investigation of Bioequiva-
lence adds that “. . . the need for narrowing the acceptance interval for both AUC
and Cmax or for AUC only should be determined on a case by case basis.” (10)
The HPFB of Canada has issued Guidance for Industry on the bioequiv-
alence requirements for critical dose drugs (37). According to this guidance,
“critical dose drugs” are defined as those drugs for which comparatively small
differences in dose or concentration lead to dose- and concentration-dependent,
serious therapeutic failures and/or serious adverse drug reactions. For these
“critical dose drugs,” the 90% CI of the relative mean AUC of the test to ref-
erence formulation should lie within 90% to 112%, according to Canada’s HPFB
guidance. In addition, the 90% CI of the relative mean Cmax of the test to ref-
erence formulation for these “critical dose drugs” should be between 80% and
125%. For “uncomplicated” drugs, Canada’s HPFB requires the point estimate of
Cmax to simply lie between 80% and 125%. These requirements for “critical dose
drugs” are to be met in both the fasted and fed states. In an appendix to this
HPFB guidance a list of 9 “critical dose drugs” is given (37). The FDA Guidance
for Industry on Bioavailability and Bioequivalence Studies for Orally Adminis-
tered Drug Products recommends that the usual BE limit of 80% to 125% for non-
NTI drugs remain unchanged for the bioavailability measures (AUC and Cmax )
for NTI drug substances unless otherwise indicated by a specific guidance (34).
On the other hand, wider BE limits for the 90% CI may be acceptable for
Cmax (in certain cases) and for AUC (in rare cases) according to the EMEA Note
for Guidance (7). Indeed, when the intrasubject variability in AUC and Cmax is
high the estimated 90% CI is wide and it is very difficult to be entirely located
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
within the usually accepted BE limits of 0.80 to 1.25. Among the methods pro-
posed during recent years in the scientific literature to evaluate the BE of these
highly variable drugs and drug products, scaled average BE and expanding the
usual BE limits to, for example, 0.75 to 1.33, were recently shown to be sensitive
to differences between means and, consequently, highly effective for assessing
the equivalence of average kinetic responses (38–40). Recently, a commentary
was published in which the authors proposed to adjust the BE limits for highly
variable drugs/drug products by scaling to the intrasubject variability of the
reference product in the study (41). The recommendation for the use of reference
scaling is based on the general concept that reference variability should be used
as an index for setting the public standard expressed in the BE limit. The use
of the reference-scaling approach necessitates a study design that evaluates the
reference variability via replicate administration of the reference product to each
subject.
BE studies using a replicate design, for example a three-period or four-
period study, have certain advantages over the classical two-period design. They
allow the comparison of the intrasubject variance and the evaluation of the
subject-by-formulation interaction. Information on these variances associated
with the test and reference formulations allows assessment of the pharmaceu-
tical quality of a new test product compared to the pharmaceutical quality of the
marketed innovator product. At the moment, none of the major health authori-
ties (EMEA, FDA, HPFB), however, provide clear recommendations on how to
assess BE of highly variable drugs or drug products.
The draft version of the revised EMEA Guideline on the Investigation of
Bioequivalence is clear in this respect. According to this guideline, it is accept-
able to widen the 90% acceptance range of Cmax , but not AUC, from 0.80–1.25 to
0.75–1.33 under the following conditions: (i) the 0.75 to 1.33 acceptance range has
been prospectively defined in the study protocol, (ii) it has been prospectively
justified that widening of the acceptance criteria for Cmax does not affect clinical
efficacy or safety, and (iii) the BE study is of a replicate design where it has
been demonstrated that the intrasubject variability for Cmax of the reference
compound in the study is >30% (10).
Unlike the FDA, which has a specific guidance on SUPAC, the EMEA Note
for Guidance on the Investigation of Bioavailability and Bioequivalence only has
a small paragraph on variations:
If a product has been reformulated from the formulation initially approved
or the manufacturing method has been modified by the manufacturer in
ways that could be considered to impact on the bioavailability, a bioe-
quivalence study is required, unless otherwise justified. Any justification
presented should be based upon general considerations . . ., or on whether
an acceptable in vivo/in vitro correlation has been established. (7).
From a regulator’s and sponsor’s point of view it would be desirable to
have clear and more detailed guidelines on this important issue, such as the
SUPAC guidelines of the FDA, to guarantee the continuing quality of a generic
drug product even during the postapproval period.
document was released by the EMEA dealing more specifically with the vaso-
constriction (human skin blanching) assay that may reduce the need for data
from clinical trials when assessing therapeutic equivalence between topical cor-
ticosteroid products (60). This document refers to the FDA guidance for industry
for a detailed description of how to perform this vasoconstriction assay (61).
Although the guidance only deals with oral modified release formulations
and transdermal dosage forms, most recommendations are also applicable to
implants and intramuscular/subcutaneous depot formulations. Paragraph 5 of
this document specifically deals with applications for modified release dosage
forms essentially similar to a marketed modified release form, that is, so-called
generic applications. A distinction is made between prolonged release oral for-
mulations, delayed release oral formulations and transdermal drug delivery sys-
tems (TDDS).
curves often with multiple peaks, Cmax is of limited value to characterize the rate
of absorption. Therefore, other measures such as the half-value duration, peak-
trough fluctuation (PTF), and percent swing may be useful alternatives (63,64).
Any widening of the usual 0.80 to 1.25 acceptance criterion should be prospec-
tively established in the study protocol and should be clinically justified.
REFERENCES
1. Tempest B. India: A global strategic asset for developed world market businesses.
J Generic Med 2006; 4:37–42.
2. Nation RL, Sansom LN. Bioequivalence requirements for generic products. Pharmac
Ther 1994; 62:41–55.
3. Chen M-L, Shah V, Patnaik R, et al. Bioavailability and bioequivalence: An FDA reg-
ulatory overview. Pharm Res 2001; 18:1645–1650.
4. EC Notice to Applicants, volume 2A: Procedures for marketing authorisation. The
Rules Governing Medicinal Products in the European Union, Chapter 1: Marketing
Authorisation, November 2005. http://ec.europa.eu/enterprise/pharmaceuticals/
eudralex/ vol-2/a/vol2a chap1 2005–11.pdf. Accessed May 2009.
5. CPMP Note for Guidance on the Investigation of Bioavailability and Bioequivalence,
Brussels, December 1991.
6. Pharmacokinetic studies in man, 1987. http://www.emea.europa.eu/pdfs/human/
ewp/3cc3aen.pdf. Accessed May 2009.
7. EMEA Note for Guidance on the Investigation of Bioavailability and Bioequivalence.
London: CPMP/EWP/QWP/EMEA, July 2001. http://www.emea.europa.eu/pdfs/
human/qwp/140198enfin.pdf. Accessed May 2009.
8. EMEA Note for Guidance on Modified Release Oral and Transdermal Dosage Forms:
Section II (Pharmacokinetic and Clinical Evaluation). London: CPMP/EWP/280/96,
July 1999. http://www.emea.europa.eu/pdfs/human/ewp/028096en.pdf. Accessed
May 2009.
9. Questions & Answers on the Bioavailability and Bioequivalence Guideline.
EMEA, London: EMEA/CHMP/EWP/40326/2006, July 26, 2006. http://www.emea.
europa.eu/pdfs/human/ewp/4032606en.pdf. Accessed May 2009.
10. EMEA Guideline on the Investigation of Bioequivalence, EMEA. London:
CPMP/EWP/QWP/1401/98, July 24, 2008. Rev. 1. http://www.emea.europa.eu/
pdfs/human/qwp/140198enrev1.pdf. Accessed May 2009.
11. Verbeeck RK, Kanfer I, Walker RB. Generic substitution—the use of medicinal prod-
ucts containing different salts and implications for safety and efficacy. Eur J Pharm Sci
2006; 28:1–6.
12. Schuirmann DJ. Design of bioavailability/bioequivalence studies. Drug Inf J 1990;
24:315–323.
13. Pidgen AW. Statistical aspects of bioequivalence—A review. Xenobiotica 1992; 22:881–
893.
14. Bolton S, Bon C. Statistical considerations—Alternative designs and approaches
for bioequivalence assessments. In: Kanfer I, Shargel L, eds. Generic Product
Development—Bioequivalence Issues. New York: Informa Healthcare, 2008:123–141.
15. Diletti E, Hauschke D, Steinijans VW. Sample size determination for bioequivalence
assessment by means of confidence intervals. Int J Clin Pharmacol Ther Toxicol 1992;
30(suppl 1):S51–S58.
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
16. Chow S-C, Wang H. On sample size calculation in bioequivalence trials. J Pharma-
cokinet Pharmacodyn 2001; 28:155–169.
17. Diletti E, Hauschke D, Steinijans VW. Sample size determination: Extended tables for
the multiplicative model and bioequivalence ranges of 0.9 to 1.11 and 0.7 to 1.43. Int J
Clin Pharmacol Ther Toxicol 1992; 30(suppl 1):S59–S62.
18. HPFB Guidance for Industry. Conduct and Analysis of Bioavailability and Bioequiv-
alence Studies—Part A: Oral Dosage Formulations used for Systemic Effects. Ottawa,
1992. http://www.hc-sc.gc.ca.
19. Olsson B, Szamosi J. Multiple dose pharmacokinetics of a new once daily extended
release tolterodine formulation versus immediate release tolterodine. Clin Pharma-
cokinet 2001; 40:227–235.
20. FDA Guidance for Industry: Immediate Release Solid Oral Dosage Forms. Scale-
up and Postapproval Changes, Chemistry, Manufacturing, and Controls, In Vitro
Dissolution Testing, and In Vivo Bioequivalence Documentation. Washington,
DC: CDER/FDA, November 1995. http://www.fda.gov/cder/guidance/index.htm.
Accessed May 2009.
21. Amidon GL, Lennernas H, Shah VP, et al. A theoretical basis for a biopharmaceutic
drug classification: The correlation of in vitro drug product dissolution and in vivo
bioavailability. Pharm Res 1995; 12:413–420.
22. Polli JE. In vitro-in vivo relationship of several “immediate” release tablets containing
a low permeability drug. Adv Exp Med Biol 1997; 423:191–198.
23. Hauschke D, Steinijans V, Pigeot I. Metrics to characterize concentration-time pro-
files in single- and multiple-dose bioequivalence studies. In: Hauschke D, Steinijans
V, Pigeot I, eds. Bioequivalence Studies in Drug Development—Methods and Appli-
cations. Chichester, West Sussex, UK: Wiley, 2007:17–36.
24. Shah SA, Rathod IS, Savale SS, et al. Determination of bioequivalence of lome-
floxacin tablets using urinary excretion data. J Pharm Biomed Anal 2002; 30:1319–
1329.
25. Midha KK, Hubbard JW, Rawson M, et al. The application of partial areas in assess-
ment of rate and extent of absorption in bioequivalence studies of conventional
release products: Experimental evidence. Eur J Pharm Sci 1994; 2:351–363.
26. Endrenyi L, Tothfalusi L. Truncated AUC evaluates efectively the bioequivalence of
drugs with long half-lives. Int J Clin Pharmacol Ther 1997; 35:142–150.
27. Gaudreault J, Potvin D, Lavigne J, et al. Truncated area under the curve as a measure
of relative extent of bioavailability evaluation using experimental data and Monte
Carlo simulation. Pharm Res 1998; 15:1621–1629.
28. Sathe P, Venitz J, Lesko L. Evaluation of truncated areas in the assessment of bioequiv-
alence of immediate release formulations of drugs with long half-lives and of Cmax
with different dissolution rates. Pharm Res 1999; 16:939–943.
29. Chen M-L, Lesko L, Williams RL. Measures of exposure versus measures of rate and
extent of absorption. Clin Pharmacokinet 2001; 40:565–572.
30. HPFB Guidance for Industry: Conduct and Analysis of Bioavailability and Bioequiv-
alence Studies—Part C: Report on Bioavailability of Oral Dosage Formulations, not in
Modified Release Form, of Drugs used for Systemic Effects, having Complicated or
Variable Pharmacokinetics. Ottawa, 1992. http://www.hc-sc.gc.ca.
31. Jackson AJ, Robbie G, Marroum P. Metabolites and bioequivalence: Past en present.
Clin Pharmacokinet 2004; 43:655–672.
32. Midha KK, Rawson MJ, Hubbard JW. The role of metabolites in bioequivalence.
Pharm Res 2004; 21:1331–1344.
33. Jackson A. Role of metabolites in bioequivalence assessment. In: Kanfer I, Shargel
L, eds. Generic Product Development—Bioequivalence Issues. New York: Informa
Healthcare, 2008:171–183.
34. FDA Guidance for Industry: Bioavailability and Bioequivalence Studies for
Orally Administered Drug Products—General Considerations. Washington,
DC: CDER/FDA, March 2003. http://www.fda.gov/cder/guidance/index.htm.
Accessed May 2009.
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
35. Schuirmann DJ. A comparison of the two one-sided tests procedure and the power
approach for assessing the equivalence of average bioavailability. J Pharmacokinet
Biopharm 1987; 15:657–680.
36. Williams RL. Bioequivalence and therapeutic equivalence. In: Welling PG, Tse FLS,
Dighe SV, eds. Pharmaceutical Bioequivalence. New York: Marcel Dekker Inc., 1991:1–
15.
37. HPFB Guidance for Industry. Bioequivalence requirements: Critical dose drugs,
Ottawa, June 2006. http://www.hc-sc.gc.ca/hpfb-dgpsa/tpd-dpt. Accessed May
2009.
38. Shah VP, Yacobi A, Barr WH, et al. Evaluation of orally administered highly variable
drugs and drug formulations. Pharm Res 1996; 13:1590–1594.
39. Midha KK, Rawson MJ, Hubbard JW. Individual and average bioequivalence of
highly variable drugs and drug products. J Pharm Sci 1997; 86:1193–1197.
40. Tothfalusi L, Endrenyi L, Midha KK, et al. Evaluation of the bioequivalence of highly-
variable drugs and drug products. Pharm Res 2001; 18:728–733.
41. Haidar SH, Davit B, Chen M-L, et al. Bioequivalence approaches for highly variable
drugs and drug products. Pharm Res 2008; 25:237–241.
42. Yu LX, Amidon GL, Polli JE, et al. Biopharmaceutics classification system: The scien-
tific basis for biowaiver extensions. Pharm Res 2002; 19:921–925.
43. Polli JE, Yu LX, Cook JA, et al. Summary workshop report: Biopharmaceutics classifi-
cation system—implementation challenges and extension opportunities. J Pharm Sci
2004; 93:1375–1381.
44. FDA Guidance for Industry. Waiver of In Vivo Bioavailability and Bioequiva-
lence Studies for Immediate Release Solid Oral Dosage Forms Based on a Bio-
pharmaceutics Classification System. Washington, DC: CDER/FDA, August 2000.
http://www.fda.gov/cder/guidance/index.htm. Accessed May 2009.
45. Alt A, Potthast H, Moessinger J, et al. Biopharmaceutical characterization of sotalol-
containing oral immediate release drug products. Eur J Pharm Biopharm 2004; 58:145–
150.
46. EMEA Concept Paper on BCS-Biowaiver. EMEA/CHMP/EWP/213035/2007,
May, 24, 2007. http://www.emea.europa.eu/pdfs/human/ewp/21303507en.pdf.
Accessed May 2009.
47. Blume HH, Schug BS. The biopharmaceutics classification system (BCS): Class III
drugs—better candidates for BA/BE waiver? Eur J Pharm Sci 1999; 9:117–121.
48. Polli JE, Yu LX, Cook JA, et al. Summary workshop report: Biopharmaceutics Classifi-
cation System—implementation challenges and extension opportunities. J Pharm Sci
2004; 93:1375–1381.
49. Jantratid E, Prakongpan S, Amidon GL, et al. Feasibility of biowaiver extension to
biopharmaceutics classification system III drug products—cimetidine. Clin Pharma-
cokinet 2006; 45:385–399.
50. Wu C-Y, Benet LZ. Predicting drug disposition via application of BCS: Transport/
absorption/elimination interplay and development of a biopharmaceutics drug dis-
position classification system. Pharm Res 2005; 22:11–23.
51. The Rules Govering Medicinal Products in the European Union, Volume 2: Notice
to Applicants and Regulatory Guidelines for Medicinal Products for Human Use.
http://ec.europa.eu/enterprise/pharmaceuticals/eudralex/vol2 en.htm. Accessed
May 2009.
52. EC Notice to Applicants, volume 2C: Regulatory Guidelines—Guideline on Dossier
Requirements for Type IA and IB Notifications, July 2006. http://ec.europa.eu/
enterprise/pharmaceuticals/eudralex.
53. Karim A. Enantioselective assays in comparatve bioavailability studies of racemic
drug formulations: Nice to know or need to know? J Clin Pharmacol 1996; 36:490–
499.
54. Mehvar R, Jamali F. Bioequivalence of chiral drugs: Stereospecific versus non-
stereospecific methods. Clin Pharmacokinet 1997; 33:122–141.
SPH SPH
IHBK055-05 IHBK055-Kanfer January 8, 2010 19:41 Char Count=
55. Boni JP, Korth-Bradley JM, Richards LS, et al. Chiral Bioequivalence: Effect of absorp-
tion rate on racemic etodolac. Clin Pharmacokinet 2000; 39:459–469.
56. Mehvar R, Jamali F. Implications of chirality for assessment of bioequivalence. In:
Kanfer I, Shargel L, eds. Generic Product Development—Bioequivalence Issues. New
York: Informa Healthcare, 2008:185–206.
57. EMEA Note for Guidance on the Clinical Requirements for Locally Applied, Locally
Acting Products Containing Known Constituents. London: CPMP/EWP/239/95,
1995. http://www.emea.europa.eu/pdfs/human/ewp/023995en.pdf. Accessed May
2009.
58. EMEA Draft Guideline on the Requirements for Clinical Documentation for
Orally Inhaled Products (OIP) including the Requirements for Demonstration of
Therapeutic Equivalence between two Inhaled Products for Use in the Treat-
ment of Asthma and Chronic Obstructive Pulmonary Disease (COPD). London:
CPMP/EWP/4151/00 Rev. 1, October 18, 2007. http://www.emea.europa.eu/pdfs/
human/ewp/415100enrev1.pdf. Accessed May 2009.
59. Clinical Investigation of Corticosteroids Intended for Use on the Skin, August 1987.
http://www.emea.europa.eu/pfds/human/ewp/3cc26aen.pdf. Accessed May 2009.
60. EMEA Questions and Answer on Guideline Title: Clinical Investigation of Cor-
ticosteroids Intended for Use on the Skin. CHMP/EWP/21441/2006, November
16, 2006. http://www.emea.europa.eu/pdfs/human/ewp/2144106en.pdf. Accessed
May 2009.
61. FDA Guidance for Industry. Topical Dermatologic Corticosteroids—In Vivo Bioequiv-
alence. Washington, June 2, 1995. http://www.fda.gov/ohrms/dockets/dockets/
04p0206/04p-0206-ref0001–08-FDA-Guidance-for-Industry-06–1995-vol3.pdf.
Accessed May 2009.
62. HPFB Guidance for Industry. Conduct and Analysis of Bioavailability and Bioe-
quivalence Studies—Part B: Oral Modified Release Formulations. Ottawa, 1996.
http://www.hc-sc.gc.ca. Accessed May 2009.
63. Steinijans VW, Sauter R, Hauschke D, et al. Metrics to characterize concentration-time
profiles in single- and multiple-dose bioequivalence studies. Drug Inf J 1995; 29:981–
987.
64. Schultz H-U, Steinijans VW. Striving for standards in bioequivalence assessment: A
review. Int J Clin Pharmacol Ther Toxicol 1992; 30(suppl 1):S1–S6.
65. FDA Guidance for Industry. Skin Irritation and Sensitization Testing of Generic
Transdermal Drug Products. Washington, DC: CDER/FDA, December 1999.
http://www.fda.gov/cder/guidance/index.htm. Accessed May 2009.
66. EMEA Questions & Answers document on the clinical development of fixed combi-
nations of drugs belonging to different therapeutic classes in the field of cardiovas-
cular treatment and prevention. London: EMEA/CHMP/EWP/191583/2005, June
2005. http://www.emea.europa.eu/pdfs/human/ewp/19158305en.pdf. Accessed
May 2009.
67. Multisource (generic) pharmaceutical products: Guidelines on registration require-
ments to establish interchangeability. WHO Technical Report Series No. 937, 2006.
68. Gupta E, Barends DM, Yamashita E, et al. Review of global regulations concerning
biowaivers for immediate release solid oral dosage forms. Eur J Pharm Sci 2006;
29:315–324.
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
6 India
Subhash C. Mandal
Directorate of Drugs Control, “NALANDA,” Fartabad, Amtola, Kolkata, India
S. Ravisankar
GVK Biosciences Pvt. Ltd., Ameerpet, Hyderabad, India
INTRODUCTION
Although the concept of bioavailability (BA) and bioequivalence (BE) has been
developed over the decades and such considerations are in practice in different
research centers in academic institutions and industries in India for quite some
time, BE testing was not made mandatory by Drug Regulatory Agencies, until
the 1980s. The introduction of dissolution tests for seven products, chlorpro-
mazine, digitoxin, digoxin, lithium carbonate, quinidine, tetracycline, and
tolbutamide in the Indian Pharmacopoeia in 1985 (1), initiated the process of
framing legislation for regulatory requirements of BE studies. After this phase
(three years), BE studies became mandatory for all new drugs introduced on the
markets in India, by incorporating Schedule Y of the Drugs and Cosmetics Act
in 1988, followed by subsequent amendments of Schedule Y in 1989 and 2005
(2). The term “new drug” in India is defined under 122E of the Rule and includes
both brand and generic.
India has a large number of BA and BE centers, mostly owned and operated
by contract research organizations (CROs), largely in the private sector. More
than 950 new drugs were approved between 1988 and July 2007 in India (3),
following successful completion of necessary trials including BE and BA studies.
1. Product patents have been extended to pharmaceuticals, which was not pre-
viously permitted as per the Patent Act of 1970.
2. Patent protection has been extended to 20 years from 7 years.
3. The Indian Act denied granting of a patent retrospective to the mailing date
of the submission. Thus Indian companies that had been producing and
selling patented medicines could not be subjected to patent infringement
claims. The Act also provided that even after a patent had been granted,
Indian companies would be able to continue production subject to payment
of reasonable royalties to the patent holders. Subsection 2 of Section 5 of the
act specifies the term “reasonable,” but no explanation is given.
114
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 115
4. A most significant feature of the Act is that it provides protection from sec-
ondary patenting of the same chemical/pharmaceutical molecule. It forbids
patenting of “salts, esters, polymorphs, metabolites, pure forms, particle size,
isomers, mixtures of isomers, complexes, combinations and other derivatives
of known substances unless they differ significantly in properties with regard
to efficacy.”
5. The Act also provides for pre- and postpatent objection of patents and
describes 11 areas where one can raise objections on the granting of a patent.
This includes objection to a patent based upon existing knowledge in the pub-
lic domain (prior art). This holds good for domestic inventions and also for
imported materials and according to the following “That if the invention so
far as claimed in any claim of the complete specification was publicly known
or publicly used in India before the priority date of the claim” [Section 25 (d)]
such an application is not patentable.
6. The Act has now made a provision under Section 107 A (b), for the import
of patented commodities from any part of the world, where it is cheaper,
even though it is patented in India. This is known as parallel import. For
this purpose, it will also not be required to obtain any authorization from
the patentee. The Act simply says that “who is duly authorized under law to
produce and sell or distribute the product” will become the source for Indian
importers.
7. One of the most important areas of the Act is its provisions for compulsory
licensing. The act clearly directs that a “‘Compulsory’ license is granted with
a predominant purpose of supply in the Indian market and that the licensee
may also export the patented product.” The license shall also be granted to
remedy a practice determined after judicial or administrative process to be
anticompetitive. This particular clause may be carefully used to control exor-
bitantly high prices of patented products.
Generic Medicines
In India, generic medicines are those that are labeled with their generic names.
There is no separate law for registering generic medicines. Drugs and drug prod-
ucts are classified as either (i) “new drugs” or (ii) drugs other than new drugs.
However, in general “generic drugs” mean those that are no longer subject to
patent protection and are being marketed by their generic name. To understand
the legislation the concept of new drugs should be clear.
According to Section 122E of Drugs and Cosmetics Act new drugs are
defined as the following:
(a) A drug, as defined in the Act including bulk drug substance that has not
been used in the country to any significant extent (no clarification provided
by the Act—it is the prerogative of the licensing authority) under the condi-
tions prescribed, recommended or suggested in the labeling thereof and has
not been recognized as effective and safe by the licensing authority under
Rule 21 for the purposes claimed; provided that the limited use, if any, has
been with the permission of the licensing authority.
(b) A drug already approved by the licensing authority mentioned in Rule 21
for certain claims, which are now proposed to be marketed with modified or
new claims, namely, indications, dosage, dosage form (including sustained
release dosage form), and route of administration.
(c) A fixed dose combination of two or more drugs, individually approved ear-
lier for certain claims, which are now proposed to be combined for the first
time in a fixed ratio, or if the ratio of ingredients in an already-marketed
combination is proposed to be changed, with certain claims, viz., indica-
tions, dosage, dosage form (including sustained release dosage form) and
route of administration.
It also explained that all vaccines shall be new drugs unless certified other-
wise by the licensing authority under Rule 21 and a new drug shall continue to
be considered as a new drug for a period of four years from the date of its first
approval or its inclusion in the Indian Pharmacopoeia, whichever is earlier (6).
All new drugs are required to comply with the provisions and require-
ments of Schedule Y for registration in India. However some relaxation has been
granted for fixed dose combinations (FDC) to be registered in India (vide Annex-
ure VI of Schedule Y). For this purpose fixed dose combinations are categorized
into the following four groups:
(a) The first group of FDC includes those in which one or more of the active
ingredients are a new drug. Such FDC are treated in the same way as any
other new drug, for both clinical trials and marketing permission.
(b) The second group of FDC includes those in which the active ingredients
are already approved and marketed individually and are combined for the
first time, for a particular claim and where the ingredients are likely to have
significant interaction of a pharmacodynamic or pharmacokinetic nature
For permission to carry out clinical trials with such FDCs, a summary of
available pharmacological, toxicological, and clinical data on the individ-
ual ingredients should be submitted, along with the rationale for combining
them in the proposed ratio. In addition, acute toxicity data (LD 50) and phar-
macological data should also be submitted on the individual ingredients as
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 117
well as their combination in the proposed ratio. If clinical trials have been
carried out with the FDC in other countries, reports of such trials should be
submitted. If the FDC is marketed abroad, the regulatory trials performed
should be stated.
For marketing permission, the reports of clinical trials carried out with
the FDC in India should be submitted. The nature of the trials depends on
the claims to be made and the data already available.
(c) The third group of FDCs includes those that have already been marketed,
but in which it is proposed either to change the ratio of active ingredients or
to make a new therapeutic claim. For such FDCs, the therapeutic rationale
should be submitted to obtain permission for clinical trials and the reports of
trials should be submitted to obtain marketing permission. The nature of the
trials will depend on the claims to be made and the data already available.
(d) The fourth group of FDCs includes those whose individual active ingre-
dients have been widely used for a particular indication for years, their
concomitant use is often necessary and no claim is proposed to be made
other than convenience. Also, it must be a stable acceptable dosage form
and whose ingredients are unlikely to have significant interaction of a phar-
macodynamic or pharmacokinetic nature.
No additional animal or human data are generally required for those
FDCs, and marketing permission may be granted if the FDC has an accept-
able rationale.
If any drug does not fall under the ambit of a new drug, licenses could be
issued by the state licensing authority. India has a two-tier regulatory system.
One is Central Drugs Standard Control Organization (CDSCO) under the Gov-
ernment of India having certain powers vested in them and each state has its own
drug regulatory system having certain powers. CDSCO is authorized to approve
new drugs as defined under Rule 122E but the state drugs control agency can-
not grant new drug licenses. Therefore it is clear that a BA/BE study is needed
for manufacturing and import of new drugs, where the relevant product is com-
pared against the innovator product.
Several different provisions and sections of the Drugs and Cosmetics Rules
have been modified and amended over the years. A new provision has been
introduced for the registration of manufacturing premises of foreign drug manu-
facturers and for drug products, before their import to India. These amendments
have been in force since January 1, 2003. The notification also introduced addi-
tional provisions, some of which are as follows:
1. An increase in import license fees.
2. Increased validity period of license.
3. Deletion of the clause of exemption relating to the requirement of import
license for bulk drug for actual users.
4. A minimum requirement of 60% of retained shelf life for imported drug.
5. Conditions for the importation of small amounts of new drug products by
Government hospitals for treating their own patients.
Under the existing regulations, foreign manufacturers must apply for reg-
istration certificates for both their manufacturing premises and for individual
drugs to be imported. The applications for these registrations can be filed by the
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
authorized agents of foreign firms in India, along with the documents required
to be accompanied by the registration application, as specified in the relevant
amendments. The registration certificates are valid for three years from the date
from which they were issued.
A fee of US $1500 is charged for the registration of overseas manufacturer’s
premises and a fee of US $1000 is charged for each individual drug. The rules
now provide for inspection of the premises of a foreign manufacturer by Indian
Drug Authorities, whenever so required. In such cases, an additional fee of US
$5000 is charged. The rules also provide for payment of testing charges by reg-
istration holders. The foreign manufacturer or his authorized agent in India is
liable to report any change in the manufacturing and testing process of a drug.
However, no registration certificate is required for an inactive bulk substance
used as a pharmaceutical aid for the manufacture of the drug formulation. Regis-
tration may be suspended or cancelled in the event of any violation of the condi-
tions of registration. The new registration and import license scheme also covers
diagnostic kits, viz. HIV I and II, hepatitis B surface antigen, and blood group
detection reagents.
According to these rules, an import license is required for all types of drugs.
An import license application submitted on Form 10 is granted after the comple-
tion of the registration of overseas manufacturers and their specific drug prod-
uct(s) to be imported. The import licensee for specific drugs is valid for three
years from the date on which these were granted. The import license fee has
been fixed at Rs. 1000 for a single drug and at the rate of Rs. 100 for any addi-
tional drug. The fee for “import licenses for test and analysis” of a drug has been
kept at Rs. 100 for a single drug and at the rate of Rs. 50 for each additional drug.
Exemption from import licenses for the import of bulk drugs by the formulators
for actual use under Schedule D has been deleted. A provision has been made
that only drugs with a minimum of 60% of retained shelf life will be allowed
to be imported in the country, for example, if a drug product has a shelf life of
30 months, it will not be allowed to be imported into India more than 12 months
after manufacture.
Separate provision has been made to enable the Government hospitals to
import small quantities of essential new drugs for the treatment of their own
patients. The fee for such import licenses has been kept at Rs. 100 for a single
drug and at the rate of Rs. 50 for each additional drug.
The following guidelines describe the requirements of an applica-
tion for permission to import and/or manufacture new drugs for sale
or to conduct clinical trials as per the Drugs and Cosmetics Rules (2):
India 119
India 121
Study Design
A BE study should be designed in a manner that any formulation effects can
be distinguished from other effects. Typically, if two formulations (one test and
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
Subjects
Number of Subjects
The number of subjects required for a BE study is determined by considerations
such as the error variance from pilot study data, the expected deviation of the
test product from the reference product, significance level (usually 0.05) and the
power of the study (should be more than 80%). The minimum number of subjects
should not be less than 16, unless justified for ethical reasons. Additional subjects
can be recruited to allow for possible dropouts or removal from the study. The
withdrawn/dropout subjects can be replaced by a substitute (standby) provided
that a substitute subject follows all the study requirements.
Sequential or add-on studies are acceptable in specific cases, for example,
where a large number of subjects are required or where the results of the study
do not convey adequate statistical significance. In all cases, the final statistical
analysis must include data from all subjects, and reasons for not including partial
data as well as the non–included data must be documented in the final report.
Subject Selection
The selection of subjects and standardization is important to minimize intra- and
interindividual variations. The studies should be carried out in healthy male or
female volunteers. The choice of gender should be consistent with usage and
safety criteria. Women of child-bearing potential should be required to give
assurance that they are neither pregnant nor likely to become pregnant until after
the study. This is confirmed by pregnancy tests immediately prior to the first and
last dose of the study. Women taking contraceptive drugs should not normally be
included in the studies. If the drug product is intended to be used in both sexes,
attempts should be made to include similar proportions of males and females
in the studies. The choice of subject may be narrowed when a drug represents a
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 123
potential hazard in one group of users, for example, studies on teratogenic drugs
should be conducted only on males.
For a drug product to be used in a geriatric population, subjects of 60 years
of age or older should be included.
Volunteers are screened for suitability by means of a comprehensive med-
ical examination including clinical laboratory tests, extensive review of medical
history, use of oral contraceptives, alcohol intake, smoking, and use of drugs of
abuse. Additional medical investigations may be required before, during, and
after the study, based upon the therapeutic class of drug and safety profile.
Phenotyping/Genotyping
While designing a study protocol, adequate care should be taken to consider
pharmacogenomic issues in the context of the Indian population. Phenotyping
and/or genotyping of subjects should be considered for exploratory BA studies
and all studies using parallel group design. It may also be considered in cross-
over studies for safety or pharmacokinetic reasons. If a drug is known to be sub-
ject to a major genetic polymorphism, studies could be performed in panels of
subjects of known phenotype or genotype for the polymorphism in question.
sampling should continue until the AUC extrapolated from the time of the last
measured drug concentration to infinity time is only a small percentage (nor-
mally less than 20%) of the total AUC. Truncated AUC may be used when the
presence of enterohepatic recycling does not allow the terminal elimination rate
constant to be accurately calculated. There should be at least three sampling
points in the absorption phase, three to four around the projected Tmax , and four
points in the elimination phase (intervals between successive sampling points
should not be longer than the half-life of the drug, in general). The number of
points used to calculate the terminal elimination rate constant should preferably
be visually determined by inspection of a semilogarithmic plot of drug concen-
tration versus time. For urinary excretion studies, samples must be collected up
to seven or more half-lives.
Parameters To Be Investigated
BA/BE evaluations are based on the measurement of the concentrations of the
drug or its metabolite(s). Measurement of the active or inactive metabolites may
be necessary in certain situations:
(a) The concentration of the drug may be too low to be accurately measured in
the biological matrix.
(b) Unstable drugs.
(c) Drugs with a very short half-life.
(d) The API is a prodrug.
Racemates should be measured by using an achiral method. Measurement
of individual enantiomers is only recommended when
(a) the enantiomers exhibit different pharmacokinetic or pharmacodynamic
characteristics,
(b) primary efficacy/safety resides with the minor enantiomer, and
(c) at least one of the enantiomers undergoes nonlinear absorption.
Pharmacodynamic Studies
Studies in healthy volunteers or patients using pharmacodynamic parameters
may be used for establishing BE between two pharmaceutical products. These
studies may become necessary if quantitative analysis of the drug and/or
metabolite(s) in plasma or urine cannot be made with sufficient accuracy and
sensitivity. Furthermore, pharmacodynamic studies in humans are required if
measurements of drug concentrations cannot be used as surrogate end points
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 125
India 127
Study Products
The investigational medicinal product (test product) should be manufactured
from a production batch. The products should be stored as per storage condi-
tions stated on the label. Randomly selected samples from the shipment by the
manufacturer or sponsor should be used for the study. The number of units pro-
cured should be twice the number required for the tests to be carried out in vivo
as well as for the number of in vitro tests. These samples must be retained (reten-
tion samples) under the recommended storage conditions in the original con-
tainers for a period of three years after the conduct of study or one year after
the period of expiry whichever is earlier. This is to ensure the samples are repre-
sentative of the ones sent by the sponsor and used for all the tests. The reserve
samples should be stored in an area segregated from the area where testing is
conducted and with access limited to authorized personnel.
Data Analysis
The main concern in a BE assessment is to limit the consumer’s risk, that is, erro-
neously accepting BE, and also at the same time, minimize the manufacturer’s
risk, that is, erroneously rejecting BE. This is done by using appropriate statisti-
cal methods for data analysis and adequate sample size.
Statistical Analysis
The statistical procedure should be specified in the protocol. In the case of BE
studies, the procedures should lead to a decision scheme, which is symmetrical
with respect to the two formulations (i.e., leading to the same decision whether
the new formulation is compared to the reference product or the reference
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
product to the new formulation). The statistical analysis (e.g., ANOVA) should
take into account sources of variation that can be reasonably assumed to have an
effect on the response.
The 90% confidence interval (CI) for the ratio of the population means
(test/reference) and the two one-sided t test with the null hypothesis of non-
BE at the 5% significance level for the parameter under consideration are applied
for assessing BE.
To meet the assumption of normality of data underlying the statistical anal-
ysis, logarithmic transformation of the data should be carried out for the phar-
macokinetic parameters Cmax and AUC before performing statistical analysis.
The analysis of Tmax is desirable if it is clinically relevant. The parameter
Tmax should be analyzed using nonparametric methods. In addition to the above,
summary statistics such as minimum, maximum, and ratio of pharmacokinetic
parameters should be given in the report.
The study protocol should specify the methods for identifying biologically
implausible outliers. Post hoc exclusion of outliers is not recommended. A scien-
tific explanation should be provided to justify the exclusion of a volunteer from
the analysis.
Acceptance Criteria
Single-Dose Studies
To establish BE, the calculated 90% CI for AUC and Cmax should fall within the
BE range, usually 80% to 125%. This is equivalent to the rejection of the two one-
sided t test with the null hypothesis of non-BE at a 5% level of significance. The
nonparametric 90% CI for Tmax should lie within a clinically acceptable range.
Tighter limits for permissible differences in BA may be required for drugs
that have
(i) a narrow therapeutic index,
(ii) a serious, dose-related toxicity,
(iii) a steep dose/effect curve, or
(iv) nonlinear pharmacokinetics within the therapeutic dose range.
A wider acceptance range may be acceptable if it is based on sound clinical
justification.
India 129
Study Parameters
BA data should be obtained for all modified-release drug products although the
type of studies required and the pharmacokinetic parameters that should be eval-
uated may differ depending on the active ingredient involved.
Factors to be considered include whether or not the formulation represents
the first market entry of the drug substance, and the extent of accumulation of
the drug after repeated dosing.
If the formulation is the first market entry of the drug substance, the
product’s pharmacokinetic parameters should be determined. If the formula-
tion is a second or subsequent market entry then comparative BA studies using
an appropriate reference product, should be performed. For the first generic
drug product, the international innovator drug product should be used as
the reference product but for subsequent products, it may be the reference
product approved earlier, which was equivalent to the international innovator
product.
Study Design
The study design should be a single dose or single and multiple dose based
on the modified-release products that are likely to accumulate or unlikely to
accumulate, both in the fasted and nonfasting state. If the effect of food on the
reference product is not known (or it is known that food affects its absorption),
two separate two-way cross-over studies, a fasted and a fed study should be
carried out.
If it is known with certainty (e.g., from published data) that the reference
product is not affected by food, then a three-way cross-over study may be appro-
priate with
r the reference product in the fasting state,
r the test product in the fasted state, and
r the test product in the fed state.
When the modified-release product is the first market entry of that type of
dosage form, the reference product should normally be the innovator’s immedi-
ate release formulation. The comparison should be between a single dose of the
modified-release formulation and doses of the immediate-release formulation,
which it is intended to replace. The latter must be administered according to the
established dosing regimen.
When the modified-release product is the second or subsequent entry on
the market, comparison should be with the reference modified-release product
for which BE is claimed.
Studies should be performed using single-dose administration in the fast-
ing state as well as following an appropriate meal at a specified time. The fol-
lowing pharmacokinetic parameters should be calculated from plasma (or rele-
vant biological matrix) concentrations of the drug and/or major metabolite(s):
AUC0– , AUC0–t , AUC0–∞ , and Cmax (where the comparison is with an existing
modified-release product), and kel . The 90% CI calculated using log-transformed
data for the ratios (test/reference) of the geometric mean AUC (for both AUC0–
and AUC0–t ) and Cmax (where the comparison is with an existing modified-
release product) should generally be within the range 80% to 125% both in the
fasting state and following the administration of an appropriate meal at a speci-
fied time before taking the drug.
The pharmacokinetic parameters should support the claimed dose delivery
attributes of the modified-release dosage form.
India 131
The 90% CI for the ratio of geometric means (test/reference drug) for
AUC0– (ss) , Cmax , and Cmin determined using log-transformed data should gen-
erally be within the range 80% to 125% when the formulations are compared at
steady state.
The pharmacokinetic parameters should support the claimed attributes of
the modified-release dosage form. Pharmacodynamic data may reinforce or clar-
ify interpretation of differences in the plasma concentration data. Where these
studies do not show BE, comparative efficacy, and safety data may be required
for the new product.
Reporting of Results
The BE report should give complete documentation with respect to the protocol,
conduct and evaluation of the study. The report should include (as a minimum)
the following information:
4.10.1. Table of contents
4.10.2. Title of the study
4.10.3. Names and credentials of responsible investigators
4.10.4. Signatures of the principal and other responsible investigators authenti-
cating their respective sections of the report
4.10.5. Site of the study and facilities used
4.10.6. The period of dates over which the clinical and analytical steps were con-
ducted
4.10.7. Names, batch numbers, and expiry date of the products compared (for the
test product, an expiry date may not be available prior to conducting the
study)
4.10.8. A signed declaration that this product is identical to that intended for
marketing
4.10.9. Results of assays and other pharmaceutical tests (e.g., physical descrip-
tion, dimensions, mean weight, weight uniformity, and comparative disso-
lution) carried out on the batches of the various products
4.10.10. Full protocol for the study including a copy of the Informed Consent
Form and criteria for inclusion/exclusion or withdrawal of subjects
4.10.11. Report of protocol deviations, violations
4.10.12. Documentary evidence that the study was approved by an independent
ethics committee and was carried out in accordance with good clinical prac-
tice/good laboratory practice.
4.10.13. Demographic data of subjects
4.10.14. Names and addresses of subjects
4.10.15. Details of and justification for protocol deviations
4.10.16. Details of dropout and withdrawals from the study should be fully doc-
umented and accounted for
4.10.17 Details of analytical methods used, full validation data, QC data, and
criteria for accepting or rejecting assay results
4.10.18 Representative chromatograms (normally 20%) covering the whole con-
centration range for all, standard and QC samples as well as specimens
analyzed
4.10.19. Sampling schedules and deviations of the actual times from the sched-
uled times
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
Quality Assurance
Systems and processes established to ensure that the trial is performed and the
data are generated in compliance with GCP must be in place. Quality assurance
(QA) is validated through in-process QC and in- and postprocess auditing of
clinical trial process as well as data.
The sponsor is responsible for the implementation of a system of QA to
ensure that the study is performed and the data are generated, recorded, and
reported in compliance with the protocol, GCP, and other applicable require-
ments. Documented standard operating procedures (SOPs) are a prerequisite
for QA.
All observations and findings should be verifiable for the credibility of the
data and to assure that the conclusions presented are correctly derived from the
raw data. Verification processes must therefore be specified and justified.
Statistically controlled sampling may be an acceptable method of data ver-
ification in each study. QC must be applied to each stage of data handling to
ensure that all data are reliable and have been processed correctly.
Sponsor audits should be conducted by persons independent of those
responsible for the study. Investigational sites, facilities, all data and documen-
tation should be available for inspection and audit by the sponsor’s auditor as
well as by the regulatory authority (ies).
Other considerations for quality documentation are the following:
a. A meticulous and specified plan for the various steps and procedures for the
purpose of controlling and monitoring the study most effectively.
b. Specifications and instructions for anticipated deviations from the protocol.
c. Allocation of duties and responsibilities within the research team and their
coordination.
d. Instructions to staff including study description (the way the study is to be
conducted and the procedures for drug usage and administration).
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 133
e. Addresses and contact numbers, etc., enabling any staff member to contact
the research team at any hour.
f. Considerations of confidentiality problems, if any arise.
g. QC of methods and evaluation procedures.
For criteria (e) and (f), the applicant is expected to demonstrate that the
excipients in the new drug are essentially the same and in comparable concen-
trations as those in the reference product. In the event that this information about
the reference product cannot be provided by the applicant, in vivo studies need
to be performed.
In Vitro Studies
Under the following circumstances BE may be assessed by the use of in vitro
dissolution testing.
a. Drugs for which the applicant provides data to substantiate all of the
following:
i. Highest dose strength is soluble in 250 mL of an aqueous media over the
pH range of 1 to 7.5 at 37◦ C.
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
ii. At least 90% of the administered oral dose is absorbed on mass balance
determination or in comparison to an intravenous reference dose.
iii. Speed of dissolution as demonstrated by more than 80% dissolution
within 15 minutes at 37◦ C using IP apparatus 1, at 50 rpm or IP appa-
ratus 2, at 100 rpm in a volume of 900 mL or less in each of the following
media:
1. 0.1 N hydrochloric acid or artificial gastric juice (without enzymes).
2. A pH 4.4 buffer.
3. A pH 6.8 buffer or artificial intestinal juice (without enzymes).
b. Different strength of the drug manufactured by the same manufacturer,
where all of the following criteria are fulfilled:
i. The qualitative compositions between the strength is essentially the
same.
ii. The ratio of active ingredients and excipients between the strength is
essentially the same, or in the case of low strengths, the ratio between
the excipients is the same.
iii. The methods of manufacture is essentially the same.
iv. An appropriate BE study has been performed on at least one of the
strengths of the formulations (usually the highest strengths unless a
lower strength is chosen for reasons of safety).
v. In case of systemic availability—pharmacokinetics have been shown to
be linear over the therapeutic dose range.
India 135
REFERENCES
1. Pharmacopoeia of India, Vol I, 3rd ed. Ministry of Health and Family Welfare, Gov-
ernment of India, xvi, 1985.
2. The Drugs and Cosmetics Act 1940 & The Drugs and Cosmetics Rules 1945, Ministry
of Health and Family Welfare, Government of India, 425–459, 2005.
3. Ghosh A, Hazra A, Mandal SC, New Drugs in India over the past 15 years: Analysis
of trends. Natl Med J India 2004; 17(1):10–16.
4. The Patents (Amendment) Act 2005, The Gazette of India: Extraordinary, Part II-Sec.
I, 2005:1–18.
5. The Patents (Amendment) Rules, 2005, The Gazette of India: Extraordinary, Part II-
Sec. 3 (ii), 2004:60–108.
6. The Drugs and Cosmetics Act 1940 & The Drugs and Cosmetics Rules 1945, Ministry
of Health and Family Welfare, Government of India, 97, 2005.
7. Guidelines for Bioavailability & Bioequivalance Studies, Central Drugs Standard
Control Organization, Directorate General of Health Services, Ministry of Health &
Family Welfare, Government of India, New Delhi, March 9–12, 2005.
8. GCP (Good Clinical Practice) Guidelines issued by CDSCO (Central Drugs Standards
Control Organization), Ministry of Health and Family Welfare, India.
9. Schedule Y, and amendment to Drugs & Cosmetic Act & Rules, Ministry of Health
and Family Welfare, India, 2005.
10. The Ethical Guidelines for Biomedical research on human subjects issued by Indian
Council of Medical Research, India, 2006.
Enantiomeric purity
Assay
2.5. Validation:
Assay method
Impurity estimation method
Residual solvent/other volatile impurities (OVI) estimation method
2.6. Stability studies (for details refer Appendix IX)
Final release specification
Reference standard characterization
Material safety data sheet
2.7. Data on Formulation:
Dosage form
Composition
Master manufacturing formula
Details of the formulation (including inactive ingredients)
In-process quality control check
Finished product specification
Excipient compatibility study
Validation of the analytical method
Comparative evaluation with international brand(s) or approved Indian
brands, if applicable
Pack presentation
Dissolution
Assay
Impurities
Content uniformity
pH
Forced degradation studies
Stability evaluation in market intended pack at proposed storage con-
ditions
Packing specifications
Process validation
When the application is for clinical trials only, the INN or generic name,
drug category, dosage form, and data supporting stability in the intended
container-closure system for the duration of the clinical trial (information
covered in items 2.1, 2.3, 2.6, 2.7) are required.
3. Animal Pharmacology (for details refer Appendix IV)
3.1. Summary
3.2. Specific pharmacological actions
3.3. General pharmacological actions
3.4. Follow-up and Supplemental Safety Pharmacology Studies
3.5. Pharmacokinetics: absorption, distribution; metabolism; excretion
4. Animal toxicology (for details refer Appendix III)
4.1. General aspects
4.2. Systemic toxicity studies
4.3. Male fertility study
4.4. Female reproduction and developmental toxicity studies
4.5. Local toxicity
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 137
4.6. Allergenicity/hypersensitivity
4.7. Genotoxicity
4.8. Carcinogenicity
5. Human/Clinical pharmacology (Phase I)
5.1. Summary
5.2. Specific pharmacological effects
5.3. General pharmacological effects
5.4. Pharmacokinetics, absorption, distribution, metabolism, excretion
5.5. Pharmacodynamics/early measurement of drug activity
6. Therapeutic exploratory trials (Phase II)
6.1. Summary
6.2. Study report(s) as given in Appendix II
7. Therapeutic confirmatory trials (Phase III)
7.1. Summary
7.1. Individual study reports with listing of sites and investigators.
8. Special studies
8.1. Summary
8.1. Bioavailability/Bio-equivalence.
8.1. Other studies, for example, geriatrics, pediatrics, pregnant, or nursing
women
9. Regulatory status in other countries
9.1. Countries where the drug is
a. Marketed
b. Approved
c. Approved as IND
d. Withdrawn, if any, with reasons
9.2 Restrictions on use, if any, in countries where marketed /approved
9.2 Free sale certificate or certificate of analysis, as appropriate.
10. Prescribing information
10.1. Proposed full prescribing information
10.2. Drafts of labels and cartons
11. Samples and testing protocol/s
11.1. Samples of pure drug substance and finished product (an equivalent
of 50 clinical doses, or more number of clinical doses if prescribed by
the licensing authority), with testing protocol/s, full impurity profile,
and release specifications.
Notes:
(1) All items may not be applicable to all drugs. For explanation, refer to text of Schedule Y.
(2) For requirements of data to be submitted with application for clinical trials refer to the text
of this Schedule.
India 139
8. Introduction:
A brief description of the product development rationale should be given
here.
9. Study objective:
A statement describing the overall purpose of the study and the primary
and secondary objectives to be achieved should be mentioned here.
10. Investigational plan:
This section should describe the overall trial design, the subject selection cri-
teria, the treatment procedures, blinding/randomization techniques
if any, allowed/disallowed concomitant treatment, the efficacy and
safety criteria assessed, the data quality assurance procedures, and the
statistical methods planned for the analysis of the data obtained.
11. Trial subjects:
A clear accounting of all trial subjects who entered the study will be given
here. Mention should also be made of all cases that were dropouts
or protocol deviations. Enumerate the patients screened, randomized,
and prematurely discontinued. State reasons for premature discontin-
uation of therapy in each applicable case.
12. Efficacy evaluation:
The results of evaluation of all the efficacy variables will be described in
this section with appropriate tabular and graphical representation. A
brief description of the demographic characteristics of the trial patients
should also be provided along with a listing of patients and observa-
tions excluded from efficacy analysis.
13. Safety evaluation:
This section should include the complete list of all serious adverse events,
whether expected or unexpected and adverse events whether serious
or not (compiled from data received as per Appendix XI).
The comparison of adverse events across study groups may be presented in
a tabular or graphical form. This section should also give a brief nar-
rative of all important events considered related to the investigational
product.
14. Discussion and overall conclusion:
Discussion of the important conclusions derived from the trial and scope for
further development.
15. List of references
16. Appendices
List of Appendices to the Clinical Trial Report
a. Protocol and amendments
b. Specimen of Case Record Form
c. Investigators’ name(s) with contact addresses, phone, email etc.
d. Patient data listings
e. List of trial participants treated with investigational product
f. Discontinued participants
g. Protocol deviations
h. CRFs of cases involving death and life threatening adverse event cases
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
General Principles
Toxicity studies should comply with the norms of good laboratory practice
(GLP). Briefly, these studies should be performed by suitably trained and qual-
ified staff employing properly calibrated and standardized equipment of ade-
quate size and capacity. Studies should be done as per written protocols with
modifications (if any) verifiable retrospectively. Standard operating procedures
(SOPs) should be followed for all managerial and laboratory tasks related to
these studies. Test substances and test systems (in vitro or in vivo) should
be properly characterized and standardized. All documents belonging to each
study, including its approved protocol, raw data, draft report, final report, and
histology slides and paraffin tissue blocks should be preserved for a minimum
of five years after marketing of the drug.
Toxicokinetic studies (generation of pharmacokinetic data either as an inte-
gral component of the conduct of nonclinical toxicity studies or in specially
designed studies) should be conducted to assess the systemic exposure achieved
in animals and its relationship to dose level and the time course of the toxicity
study. Other objectives of toxicokinetic studies include obtaining data to relate
the exposure achieved in toxicity studies to toxicological findings and contribute
to the assessment of the relevance of these findings to clinical safety, to support
the choice of species and treatment regimen in nonclinical toxicity studies, and to
provide information, which, in conjunction with the toxicity findings, contributes
to the design of subsequent nonclinical toxicity studies.
India 141
Notes:
(i) Single-dose toxicity study: Each group should contain at least five animals of either sex. At
least four graded doses should be given. Animals should be exposed to the test substance
in a single bolus or by continuous infusion or several doses within 24 hours. Animals
should be observed for 14 days. Signs of intoxication, effect on body weight, gross patho-
logical changes should be reported. It is desirable to include histopathology of grossly
affected organs, if any.
(ii) Dose-ranging study: Objectives of this study include the identification of target organ of
toxicity and establishment of MTD for subsequent studies.
(a) Rodents: Study should be performed in one rodent species (preferably rat) by the
proposed clinical route of administration. At least four graded doses including control
should be given, and each dose group as well as the vehicle control should consist of a
minimum of five animals of each sex. Animals should be exposed to the test substance
daily for 10 consecutive days. Highest dose should be the MTD of single-dose study.
Animals should be observed daily for signs of intoxication (general appearance, activity,
and behavior, etc.), and periodically for the body weight and laboratory parameters.
Gross examination of viscera and microscopic examination of affected organs should be
done. (b) Non–rodent species: One male and one female are to be taken for ascending
Phase MTD study. Dosing should start after initial recording of cage-side and laboratory
parameters. Starting dose may be three to five times the extrapolated effective dose or
MTD (whichever is less), and dose escalation in suitable steps should be done every
third day after drawing the samples for laboratory parameters. Dose should be lowered
appropriately when clinical or laboratory evidence of toxicity is observed. Administration
of test substance should then continue for 10 days at the well-tolerated dose level fol-
lowing which, samples for laboratory parameters should be taken. Sacrifice, autopsy, and
microscopic examination of affected tissues should be performed as in the case of rodents.
(iii) 14- to 28-day repeated-dose toxicity studies: One rodent (6–10/sex/group) and one non–
rodent (2–3/sex/group) species are needed. Daily dosing by proposed clinical route at
three dose levels should be done with highest dose having observable toxicity, mid-dose
between high and low dose, and low dose. The doses should preferably be multiples of the
effective doses and free from toxicity. Observation parameters should include cage-side
observations, body weight changes, food/water intake, blood biochemistry, hematology,
and gross and microscopic studies of all viscera and tissues.
(iv) 90-day repeated-dose toxicity studies: One rodent (15–30/sex/group) and one non–rodent
(4–6/sex/group) species are needed. Daily dosing by proposed clinical route at three
graded dose levels should be done. In addition to the control a “high-dose-reversal”
group and its control group should be also included. Parameters should include signs of
intoxication (general appearance, activity, behavior, etc.), body weight, food intake, blood
biochemical parameters, hematological values, urine analysis, organ weights, gross and
microscopic study of viscera and tissues. Half the animals in “reversal” groups (treated
and control) should be sacrificed after 14 days of stopping the treatment. The remaining
animals should be sacrificed after 28 days of stopping the treatment or after the recovery
of signs and/or clinical pathological changes—whichever comes later, and evaluated for
the parameters used for the main study.
(v) 180-day repeated-dose toxicity studies: One rodent (15–30/sex/group) and one non–
rodent (4–6/sex/group) species are needed. At least four groups, including control,
should be taken. Daily dosing by proposed clinical route at three graded dose levels
should be done. Parameters should include signs of intoxication, body weight, food intake,
blood biochemistry, hematology, urine analysis, organ weights, gross and microscopic
examination of organs and tissues.
India 143
dose groups, the highest one showing minimal toxicity in systemic studies, and
a control group should be taken. Each group should consist of six adult male
animals. Animals should be treated with the test substance by the intended route
of clinical use for minimum 28 days and maximum 70 days before they are paired
with female animals of proven fertility in a ratio of 1:2 for mating.
Drug treatment of the male animals should continue during pairing. Pair-
ing should be continued till the detection of vaginal plug or 10 days, whichever
is earlier. Females thus getting pregnant should be examined for their fertility
index after day 13 of gestation. All the male animals should be sacrificed at the
end of the study. Weights of each testis and epididymis should be separately
recorded. Sperms from one epididymis should be examined for their motility
and morphology. The other epididymis and both testes should be examined for
their histology.
The control and the treated groups should consist of at least 20 pregnant
rats (or mice) and 12 rabbits, on each dose level. All fetuses should to be sub-
jected to gross examination, one of the fetuses should be examined for skeletal
abnormalities and the other half for visceral abnormalities. Observation parame-
ters should include (dames) signs of intoxication, effect on body weight, effect on
food intake, examination of uterus, ovaries and uterine contents, number of cor-
pora lutea, implantation sites, resorptions (if any); and for the fetuses, the total
number, gender, body length, weight and gross/visceral/skeletal abnormalities,
if any.
Local Toxicity
These studies (see Appendix I, item 4.5) are required when the new drug is pro-
posed to be used by some special route (other than oral) in humans. The drug
should be applied to an appropriate site (e.g., skin or vaginal mucous membrane)
to determine local effects in a suitable species. Typical study designs for these
studies should include three dose levels and untreated and/ or vehicle control,
preferably use of two species, and increasing group size with increase in duration
of treatment. Where dosing is restricted due to anatomical or humane reasons, or
the drug concentration cannot be increased beyond a certain level due to the
problems of solubility, pH or tonicity, a clear statement to this effect should be
given. If the drug is absorbed from the site of application, appropriate systemic
toxicity studies will also be required.
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 145
Notes:
(i) Dermal toxicity study: The study should be done in rabbit and rat. Daily topical (dermal)
application of test substance in its clinical dosage form should be done. Test material
should be applied on shaved skin covering not less than 10% of the total body surface
area. Porous gauze dressing should be used to hold liquid material in place. Formulations
with different concentrations (at least three) of test substance, several fold higher than
the clinical dosage form should be used. Period of application may vary from 7 to 90
days depending on the clinical duration of use. Where skin irritation is grossly visible
in the initial studies, a recovery group should be included in the subsequent repeated-
dose study. Local signs (erythema, edema, and eschar formation) as well as histological
examination of sites of application should be used for evaluation of results.
(ii) Photoallergy or dermal phototoxicity: It should be tested by Armstrong/Harber Test in
guinea pig. This test should be done if the drug or a metabolite is related to an agent
causing photosensitivity or the nature of action suggests such a potential (e.g., drugs to
be used in treatment of leucoderma). Pretest in eight animals should screen four con-
centrations (patch application for 2 hours ± 15 minutes) with and without UV expo-
sure (10 J/cm2 ). Observations recorded at 24 and 48 hours should be used to ascertain
highest nonirritant dose. Main test should be performed with 10 test animals and 5 con-
trols. Induction with the dose selected from pretest should use 0.3 mL/patch for 2 hour ±
15 minutes followed by 10 J/cm2 of UV exposure. This should be repeated on day 0, 2, 4,
7, 9, and 11 of the test. Animals should be challenged with the same concentration of test
substance between day 20 and 24 of the test with a similar 2-hour application followed
by exposure to 10 J/cm2 of UV light. Examination and grading of erythema and edema
formation at the challenge sites should be done 24 and 48 hours after the challenge. A
positive control like musk ambrett or psoralin should be used.
(iii) Vaginal toxicity test: Study is to be done in rabbit or dog. Test substance should be applied
topically (vaginal mucosa) in the form of pessary, cream, or ointment. Six to ten animals
per dose group should be taken. Higher concentrations or several daily applications of
test substance should be done to achieve multiples of daily human dose. The minimum
duration of drug treatment is seven days (more according to clinical use), subject to a
maximum of 30 days. Observation parameters should include swelling, closure of introi-
tus, and histopathology of vaginal wall.
(iv) Rectal tolerance test: For all preparations meant for rectal administration this test may be
performed in rabbits or dogs. Six to ten animals per dose group should be taken. Formu-
lation in volume comparable to human dose (or the maximum possible volume) should
be applied once or several times daily, per rectally, to achieve administration of multi-
ples of daily human dose. The minimum duration of application is seven days (more
according to clinical use), Subject to a maximum of 30 days. Size of suppositories may be
smaller, but the drug content should be several folds higher than the proposed human
dose. Observation parameters should include clinical signs (sliding on backside), signs
of pain, blood and/or mucus in feces, condition of anal region/sphincter, gross, and (if
required) histological examination of rectal mucosa.
(v) Parenteral drugs: For products meant for intravenous or intramuscular or subcutaneous
or intradermal injection the sites of injection in systemic toxicity studies should be spe-
cially examined grossly and microscopically. If needed, reversibility of adverse effects
may be determined on a case-to-case basis.
(vi) Ocular toxicity studies (for products meant for ocular instillation): These studies should
be carried out in two species, one of which should be the albino rabbit, which has a
sufficiently large conjunctival sac. Direct delivery of drug onto the cornea in case of
animals having small conjunctival sacs should be ensured. Liquids, ointments, gels, or
soft contact lenses (saturated with drug) should be used. Initial single-dose application
should be done to decide the exposure concentrations for repeated-dose studies and
the need to include a recovery group. Duration of the final study will depend on the
proposed length of human exposure subject to a maximum of 90 days. At least two
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
different concentrations exceeding the human dose should be used for demonstrating the
margin of safety. In acute studies, one eye should be used for drug administration and
the other kept as control. A separate control group should be included in repeated-dose
studies.
Slit-lamp examination should be done to detect the changes in cornea, iris, and aque-
ous humor. Fluorescent dyes (sodium fluorescein, 0.25–1.0%) should be used for detecting
the defects in surface epithelium of cornea and conjunctiva. Changes in intraocular ten-
sion should be monitored by a tonometer. Histological examination of eyes should be
done at the end of the study after fixation in Davidson’s or Zenker’s fluid.
(vii) Inhalation toxicity studies: The studies are to be undertaken in one rodent and one
non–rodent species using the formulation that is to be eventually proposed to be mar-
keted. Acute, subacute, and chronic toxicity studies should be performed according to
the intended duration of human exposure. Standard systemic toxicity study designs
(described earlier) should be used. Gases and vapors should be given in whole body
exposure chambers; aerosols are to be given by nose-only method. Exposure time and
concentrations of test substance (limit dose of 5 mg/L) should be adjusted to ensure expo-
sure at levels comparable to multiples of intended human exposure. Three dose groups
and a control (plus vehicle control, if needed) are required. Duration of exposure may
vary subject to a maximum of 6 h/d and five days a week. Food and water should be
withdrawn during the period of exposure to test substance.
Temperature, humidity, and flow rate of exposure chamber should be recorded and
reported. Evidence of exposure with test substance of particle size of 4 m (especially for
aerosols) with not less that 25% being 1 m should be provided. Effects on respiratory
rate, findings of bronchial lavage fluid examination, and histological examination of res-
piratory passages and lung tissue should be included along with the regular parameters
of systemic toxicity studies or assessment of margin of safety.
Allergenicity/Hypersensitivity
Standard tests include guinea pig maximization test (GPMT) and local lymph
node assay (LLNA) in mouse. Any one of the two may be done.
Notes:
(i) Guinea pig maximization test: The test is to be performed in two steps: first, determination
of maximum nonirritant and minimum irritant doses, and second, the main test. The initial
study will also have two components. To determine the intradermal induction dose, four
dose levels should be tested by the same route in a batch of four males and four females
(two of each sex should be given Freund’s adjuvant). The minimum irritant dose should
be used for induction. Similarly, a topical minimum irritant dose should be determined
for challenge. This should be established in two males and two females. A minimum of
six males and six females per group should be used in the main study. One test and one
control group should be used. It is preferable to have one more positive control group.
Intradermal induction (day 1) coupled with topical challenge (day 21) should be done. If
there is no response, re-challenge should be done 7 to 30 days after the primary challenge.
Erythema and edema (individual animal scores as well as maximization grading) should
be used as evaluation criteria.
(ii) Local lymph node assay: Mice used in this test should be of the same sex, either only males
or only females. Drug treatment is to be given on ear skin. Three graded doses, the highest
being maximum nonirritant dose plus vehicle control should be used. A minimum of six
mice per group should be used. Test material should be applied on ear skin on three consec-
utive days and on day 5, the draining auricular lymph nodes should be dissected out five
hours after IV 3 H-thymidine or bromo-deoxy-uridine (BrdU). Increase in 3 H-thymidine or
BrdU incorporation should be used as the criterion for evaluation of results.
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 147
Genotoxicity
Genotoxic compounds, in the absence of other data, shall be presumed to be
transspecies carcinogens, implying a hazard to humans. Such compounds need
not be subjected to long-term carcinogenicity studies. However, if such a drug is
intended to be administered for chronic illnesses or otherwise over a long period
of time—a chronic toxicity study (up to one year) may be necessary to detect
early tumorigenic effects.
Genotoxicity tests are in vitro and in vivo tests conducted to detect com-
pounds, which induce genetic damage directly or indirectly. These tests should
enable hazard identification with respect to damage to DNA and its fixation.
The following standard test battery is generally expected to be conducted:
Notes:
Ames’ Test (Reverse mutation assay in Salmonella): S. typhimurium tester strains such as TA98,
TA100, TA102, TA1535, TA97, or Escherichia coli WP2 uvrA or E. coli WP2 uvrA (pKM101) should
be used.
(i) In vitro exposure (with and without metabolic activation, S9 mix) should be done at a
minimum of five log dose levels. “Solvent” and “positive” control should be used. Posi-
tive control may include 9-amino-acridine, 2-nitrofluorine, sodium azide, and mitomycin
C, respectively, in the tester strains mentioned above. Each set should consist of at least
three replicates. A 2.5-fold (or more) increase in number of revertants in comparison to
spontaneous revertants would be considered positive.
(ii) In vitro cytogenetic assay: The desired level of toxicity for in vitro cytogenetic tests
using cell lines should be greater than 50% reduction in cell number or culture con-
fluency. For lymphocyte cultures, an inhibition of mitotic index by greater than 50% is
considered sufficient. It should be performed in CHO cells or on human lymphocyte
in culture. In vitro exposure (with and without metabolic activation, S9 mix) should be
done using a minimum of three log doses. “Solvent” and “positive” control should be
included. A positive control such as cyclophosphamide with metabolic activation and
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
mitomycin C for without metabolic activation should be used to give a reproducible and
detectable increase clastogenic effect over the background, which demonstrates the sen-
sitivity of the test system. Each set should consist of at least three replicates. Increased
number of aberrations in metaphase chromosomes should be used as the criteria for
evaluation.
(iii) In vivo micronucleus assay: One rodent species (preferably mouse) is needed. Route of
administration of test substance should be the same as intended for humans. Five ani-
mals per sex per dose groups should be used. At least three dose levels, plus “solvent”
and “positive” control should be tested. A positive control like mitomycin C or cyclophos-
phamide should be used. Dosing should be done on day 1 and 2 of study followed by
sacrifice of animals six hours after the last injection. Bone marrow from both the femora
should be taken out, flushed with fetal bovine serum (20 minutes), pelletted and smeared
on glass slides. Giemsa–MayGruenwald staining should be done and increased number
of micronuclei in polychromatic erythrocytes (minimum 1000) should be used as the eval-
uation criteria.
(iv) In vivo cytogenetic assay: One rodent species (preferably rat) is to be used. Route of
administration of test substance should be the same as intended for humans. Five ani-
mals/sex/dose groups should be used. At least three dose levels, plus “solvent” and
“positive” control should be tested. Positive control may include cyclophosphamide. Dos-
ing should be done on day 1 followed by intraperitoneal colchicine administration at 22
hours. Animals should be sacrificed two hours after colchicine administration. Bone mar-
row from both the femora should be taken out, flushed with hypotonic saline (20 minutes),
pelletted and resuspended in Carnoy’s fluid. Once again the cells should be pelletted and
dropped on clean glass slides with a Pasteur pipette. Giemsa staining should be done and
increased number of aberrations in metaphase chromosomes (minimum 100) should be
used as the evaluation criteria.
India 149
strain of animals should not have a very high or very low incidence of sponta-
neous tumors.
At least three dose levels should be used. The highest dose should be sub-
lethal, and it should not reduce the lifespan of animals by more than 10% of
expected normal. The lowest dose should be comparable to the intended human
therapeutic dose or a multiple of it, for example, 2.5 times; to make allowance for
the sensitivity of the species. The intermediate dose to be placed logarithmically
between the other two doses. An untreated control and (if indicated) a vehicle
control group should be included. The drug should be administered seven days
a week for a fraction of the lifespan comparable to the fraction of human lifespan
over which the drug is likely to be used therapeutically. Generally, the period of
dosing should be 24 months for rats and 18 months for mice.
Observations should include macroscopic changes observed at autopsy
and detailed histopathology of organs and tissues. Additional tests for carcino-
genicity (short-term bioassays, neonatal mouse assay, or tests employing trans-
genic animals) may also be done depending on their applicability on a case to
case basis.
Note:
Each dose group and concurrent control group not intended to be sacrificed early should contain
at least 50 animals of each sex. A high-dose satellite group for evaluation of pathology other than
neoplasia should contain 20 animals of each sex while the satellite control group should contain
10 animals of each sex. Observation parameters should include signs of intoxication, effect on
body weight, food intake, clinical chemistry parameters, hematology parameters, urine analysis,
organ weights, gross pathology, and detailed histopathology. Comprehensive descriptions of
benign and malignant tumor development, time of their detection, site, dimensions, histological
typing, etc. should be given.
Animal Toxicity Requirements for Clinical Trials and Marketing of a New Drug
Nonclinical toxicity testing and safety evaluation data of an IND needed for the
conduct of different phases of clinical trials
Note:
Refer Appendix III (points 1.1 through 1.7 and Tables 1.8 and 1.9) for essential features of study
designs of the nonclinical toxicity studies listed below.
India 151
General Principles
Specific and general pharmacological studies should be conducted to support
use of therapeutics in humans. In the early stages of drug development enough
information may not be available to rationally select study design for safety
assessment. In such a situation, a general approach to safety pharmacology stud-
ies can be applied. Safety pharmacology studies are studies that investigate
potential undesirable pharmacodynamic effects of a substance on physiological
functions in relation to exposure within the therapeutic range or above.
General Pharmacology
Essential Safety Pharmacology
Safety pharmacology studies need to be conducted to investigate the potential
undesirable pharmacodynamic effects of a substance on physiological functions
in relation to exposure within the therapeutic range and above. These studies
should be designed to identify undesirable pharmacodynamic properties of a
substance that may have relevance to its human safety; to evaluate adverse phar-
macodynamic and/or pathophysiological effects observed in toxicology and/or
clinical studies; and to investigate the mechanism of the adverse pharmacody-
namic effects observed and/or suspected.
The aim of the essential safety pharmacology is to study the effects of the
test drug on vital functions. Vital organ systems such as cardiovascular, respi-
ratory, and central nervous systems should be studied. Essential safety pharma-
cology studies may be excluded or supplemented based on scientific rationale.
Also, the exclusion of certain test(s) or exploration(s) of certain organs, systems
or functions should be scientifically justified.
Cardiovascular System
Effects of the investigational drug should be studied on blood pressure, heart
rate, and the electrocardiogram. If possible in vitro, in vivo, and/or ex vivo meth-
ods including electrophysiology should also be considered.
Respiratory System
Effects of the investigational drug on respiratory rate and other functions such
as tidal volume and hemoglobin oxygen saturation should be studied.
Cardiovascular System
These include ventricular contractility, vascular resistance, and the effects of
chemical mediators, their agonists and antagonists on the cardiovascular system.
Respiratory System
These include airway resistance, compliance, pulmonary arterial pressure, blood
gases, and blood pH.
Urinary System
These include urine volume, specific gravity, osmolality, pH, proteins, cytology
and blood urea nitrogen, creatinine, and plasma proteins estimation.
Gastrointestinal System
These include studies on gastric secretion, gastric pH measurement, gastric
mucosal examination, bile secretion, gastric emptying time in vivo, and ileoce-
cal contraction in vitro.
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 153
India 155
(i) I have reviewed the clinical protocol and agree that it contains all the
necessary information to conduct the study. I will not begin the study
until all necessary ethics committee and regulatory approvals have been
obtained.
(ii) I agree to conduct the study in accordance with the current protocol.
I will not implement any deviation from or changes of the protocol
without agreement by the Sponsor and prior review and documented
approval/favorable opinion from the ethics committee of the amend-
ment, except where necessary to eliminate an immediate hazard(s) to
the trial subjects or when the change(s) involved are only logistical or
administrative in nature.
(iii) I agree to personally conduct and/or supervise the clinical trial at
my site.
(iv) I agree to inform all subjects that the drugs are being used for inves-
tigational purposes and I will ensure that the requirements relating to
obtaining informed consent and ethics committee review and approval
specified in the GCP guidelines are met.
(v) I agree to report to the Sponsor all adverse experiences that occur in
the course of the investigation(s) in accordance with the regulatory and
GCP guidelines.
(vi) I have read and understood the information in the investigator’s
brochure, including the potential risks and side effects of the drug.
(vii) I agree to ensure that all associates, colleagues and employees assist-
ing in the conduct of the study are suitably qualified and experienced
and they have been informed about their obligations in meeting their
commitments in the trial.
(viii) I agree to maintain adequate and accurate records and to make those
records available for audit/inspection by the sponsor, ethics committee,
licensing authority, or their authorized representatives, in accordance
with regulatory and GCP provisions. I will fully cooperate with any
study related audit conducted by regulatory officials or authorized rep-
resentatives of the sponsor.
(ix) I agree to promptly report to the ethics committee all changes in the
clinical trial activities and all unanticipated problems involving risks to
human subjects or others.
(x) I agree to inform all unexpected serious adverse events to the sponsor
as well as the ethics committee within seven days of their occurrence.
(xi) I will maintain confidentiality of the identification of all participating
study patients and assure security and confidentiality of study data.
(xii) I agree to comply with all other requirements, guidelines, and statutory
obligations as applicable to clinical investigators participating in clinical
trials
1. Signature of investigator with date
India 157
India 159
If at any time during six months testing under the accelerated storage con-
dition, such changes that occur cause the product to fail in complying with
the prescribed standards, additional testing under an intermediate storage
condition should be conducted and evaluated against significant change
criteria.
(ii) Refrigerated study conditions for testing drug substances and formulations
(iii) Deep freeze study conditions for testing drug substances and formulations
(iv) Drug substances intended for storage below −20◦ C shall be treated on a
case-by-case basis.
(v) Stability testing of the formulation after constitution or dilution, if appli-
cable, should be conducted to provide information for the labeling on
the preparation, storage condition, and in-use period of the constituted or
diluted product. This testing should be performed on the constituted or
diluted product through the proposed in-use period.
2. Table of Contents
A complete table of contents including a list of all appendices.
i) Background and introduction
a. Preclinical experience
b. Clinical experience
Previous clinical work with the new drug should be reviewed here
and a description of how the current protocol extends existing data
should be provided. If this is an entirely new indication, how this
drug was considered for this should be discussed. Relevant infor-
mation regarding pharmacological, toxicological and other biologi-
cal properties of the drug/biological/medical device, and previous
efficacy and safety experience should be described.
ii) Study Rationale
This section should describe a brief summary of the background infor-
mation relevant to the study design and protocol methodology. The rea-
sons for performing this study in the particular population included by
the protocol should be provided.
iii) Study objective(s) (primary as well as secondary) and their logical rela-
tion to the study design.
3. Study Design
a. Overview of the study design: Including a description of the type of
study (i.e., double-blind, multicenter, placebo-controlled, etc.), a detail
of the specific treatment groups and number of study subjects in each
group and investigative site, subject number assignment, and the type,
sequence and duration of study periods.
b. Flow chart of the study
c. A brief description of the methods and procedures to be used during the
study.
d. Discussion of study design: This discussion details the rationale for the
design chosen for this study.
4. Study Population
The number of subjects required to be enrolled in the study at the investiga-
tive site and by all sites along with a brief description of the nature of the
subject population required is also mentioned.
5. Subject eligibility
a. Inclusion criteria
b. Exclusion criteria
6. Study assessments—plan, procedures and methods to be described in detail
7. Study conduct stating the types of study activities that would be included
in this section would be: medical history, type of physical examina-
tion, blood or urine testing, electrocardiogram (ECG), diagnostic test-
ing such as pulmonary function tests, symptom measurement, dispensa-
tion and retrieval of medication, subject cohort assignment, adverse event
review, etc.
Each visit should be described separately as Visit 1, Visit 2, etc.
Discontinued subjects: Describes the circumstances for subject withdrawal,
dropouts, or other reasons for discontinuation of subjects. State how
dropouts would be managed and if they would be replaced
SPH SPH
IHBK055-06 IHBK055-Kanfer January 8, 2010 16:4 Char Count=
India 161
India 163
7 Japan
Juichi Riku
Meiji Pharmaceutical University, Tokyo, Japan
INTRODUCTION
Generic drug products make up a relatively small proportion of the total pharma-
ceutical market in Japan. Generic drug products accounted for as little as 17.2%
of the market by volume in 2007 and around 6.2% by value. These figures pale in
comparison with countries such as the United States, Germany, and the United
Kingdom where generic product penetration levels are more than half of the total
pharmaceutical products volume.
Over the past few years, the Japanese government has launched a series
of initiatives designed to boost the use of generic drug products. In 2007, the
Japanese government officially announced a specific program to raise the generic
product volume-based share over the next five years from 17% to more than 30%
by the year 2012. They implemented a Japanese version of a system for generic
substitution in April 2008. Under the reforms, pharmacists are allowed to sub-
stitute the original branded product with a generic product with the patient’s
consent if the prescribing doctor does not mark the “substitution not allowed”
box on the prescription form accompanied by signature. The introduction of the
new prescription form is a significant milestone in encouraging the dispensing
of generic products by pharmacists.
However, not all doctors and pharmacists have been in favor of the generic
substitution initiatives because they remain dubious of generic drug products,
commonly questioning their quality and bioequivalence (BE) to the brand prod-
ucts. It seems that most of their lingering suspicions about generic products
stem from misconceptions and a lack of understanding about rigorous multistep
approval reviews of generic products in Japan.
(a) name
(b) ingredients and their quantities
(c) specifications and test methods
(d) manufacturing method and process
(e) storage conditions and expiration date
(f) dosage and administration
(g) indications, etc.
164
SPH SPH
IHBK055-07 IHBK055-Kanfer January 6, 2010 20:18 Char Count=
Japan 165
The quality, BE, and therapeutic equivalence of the generic drug prod-
uct are examined and reviewed for approval of the product as specified by
the MHLW ordinances subject to the “equivalence reviews.” The following data
must be included with the application for marketing approval.
Specifications and test methods
Stability
Bioequivalence
All documents and data are subject to the “compliance reviews” (1)
through written documentation and on-site inspections in accordance with
“Standard for Collections and Preparation of Approval Review Data” (2). The
MHLW may have the equivalence review of the generic drug product and com-
pliance review performed by an independent administrative organization, the
Pharmaceuticals and Medical Devices Agency (PMDA).
The generic drug product must undergo both paper and on-site good
manufacturing practices (GMP) reviews or inspections for each product before
approval, in precisely the same way as the original drug product was reviewed.
The MHLW will not grant an approval for a generic product unless that
product is the same quality as the originator drug, and both formulations are
pharmaceutically equivalent and bioequivalent to be considered therapeutically
equivalent. Therefore, the PMDA will rigorously conduct an Equivalence and
Compliance Review of the generic drug product in comparison with the origi-
nator drug product. The common technical document (CTD), comprising a set
of specifications for inclusion in the application dossier for the registration of
medicines and designed to be used across Europe, Japan, and the United States
does not necessarily apply for use with generic products. Generally, it takes
about a year on average for the generic drug product to be approved after the
time of application.
BIOEQUIVALENCE STUDIES
Historical Background
In 1971, the MHLW required submission of BE data in support of applications
for generic drug products for the first time, and the Guideline for Bioequivalence
SPH SPH
IHBK055-07 IHBK055-Kanfer January 6, 2010 20:18 Char Count=
166 Riku
Studies for Generic Drugs (1971) was released, in which large animals, such as
dogs and rabbits, could be used in BE studies, but humans were not required. In
1981, the 1971 Guideline was revised to require the use of humans in BE studies
on the grounds that animal experiments were considered unreliable to extrapo-
late the results for BE to humans. However, this 1981 Guideline did not resolve all
issues such as those related to validation, methods for statistical analysis, partial
acceptance of animal tests, etc.
In 1997, a thoroughly revised Guideline “The Guideline for Bioequivalence
Studies of Generic Products 1997” (1997 Guideline) (4) was introduced by the
MHLW. Since then, BE studies for generic drug products must be conducted
in accordance with the 1997 Guideline and follow-on guidelines. The Japanese
National Institute of Health Sciences (NIHS) has responsibility for the prepara-
tion of guidelines and has issued several statutory guidelines for BE studies of
generic drug products through the MHLW as follows:
Guidelines
(a) Guideline for Bioequivalence Studies of Generic Products, 1997 (4)
Oral immediate-release (conventional) dosage forms
Enteric-coated products
Oral controlled-release dosage forms
Other dosage forms
Dosage forms exempt from BE studies
(b) Guideline for Bioequivalence Studies for Different Strengths of Oral Solid
Dosage Forms, 2000 (5).
Different strengths of oral solid dosage forms with the same dosage and
administration as previously approved products, for example, 5 mg
tablet as well as a 10 mg tablet
(c) Guideline for Bioequivalence Studies for Additional Dosage Forms of Oral
Solid Dosage Forms, 2001 (6).
Additional dosage forms of oral solid dosage forms with the same dosage
and administration as previously approved products, for example,
addition of 5 mg capsule to 5 mg tablet.
(d) Guideline for Bioequivalence Studies of Generic Products for Topical
Dermal Application, 2003 (7).
Generic products for topical dermal application without systemic
action, such as ointments, skin patches, etc., including steroids, and
others.
(e) Guideline for Bioequivalence Studies for Formulation Changes of Oral Solid
Dosage Forms, 2000 (8).
Postapproval changes in the components and composition of oral solid
dosage forms other than the active ingredients.
(f) Draft Guideline for Bioequivalence Studies for Changes in Manufacturing
of Oral Solid Dosage Forms: Immediate-release (Conventional) and Enteric-
Coated Products, 2002 (9).
Postapproval changes in manufacturing of oral solid dosage forms.
This draft guideline has not been officially implemented as of November,
2009.
SPH SPH
IHBK055-07 IHBK055-Kanfer January 6, 2010 20:18 Char Count=
Japan 167
168 Riku
Dissolution test
No
No
Yes
Is there a significant difference*
in dissolution at a neutral pH ?
No
FIGURE 1 Bioequivalence test for oral immediate-release dosage forms and enteric-coated
products.
Bioequivalence test
Yes
Yes
Yes
Add-on study No
Sample size n >30
Mean Cmax, AUC:
90-111% No No
(Point estimate) Yes
90% confidence
Yes interval: 80-125% Mean Cmax, AUC:
90-111% No
(Point estimate)
Similar or Equivalent No Yes
dissolution Yes
Yes
Similar or Equivalent No
Yes
Bioequivalent dissolution
Not bioequivalent
Japan 169
Is there a significant
difference between the
Yes Other than
generic and innovator drugs
in size, shape, density and bioequivalence test
release mechanism?
No
Dissolution testing
Yes
Normal subjects
170 Riku
Test Methods
Choice of Reference and Test Products
Dissolution tests, the details of which are hereinafter described in “Dissolution
Tests” section (vide infra) should be performed on three lots of the innova-
tor product available on the domestic Japanese market to select an appropri-
ate reference product using the Japanese Pharmacopoeial (12) paddle method at
50 rpm. Among the three lots, the one which shows intermediate dissolution rate
patterns amongst the lots tested should be selected as the reference product. The
generic drug product (test) should be taken from the “Biobatch” lot, produced on
an industrial scale. This batch should be at least 1/10th or larger of the intended
production batch. A difference in drug content or potency between the test and
reference products should be less than 5%.
Study Design
In principle, cross-over studies should be employed with random assignment of
individual subjects to each group. Parallel designs can be employed for a drug
with a long half-life, for example, approximately 72 hours and longer.
Number of Subjects
A sufficient number of subjects for assessing BE should be included but a min-
imum number has not been specified in the guidelines. If BE cannot be demon-
strated because of an insufficient number, an add-on study can be performed
using not less than half the number of subjects in the initial study. Provision for
an add-on must be clearly stated a priori in the study protocol and if undertaken,
no further add-ons are subsequently permissible.
Selection of Subjects
Healthy adult human subjects (volunteers) should be employed, usually male
in practice, although not specified in the guidelines. When the test and ref-
erence products show a significant difference in dissolution around pH 6.8,
subjects with low gastric acidity (achlorhydric subjects) should be employed
unless the application of the drug is limited to a special population. Since
achlorhydria is quite common in Japan, and significant differences in BE may
not be shown in subjects with normal gastric acidity, it is important to confirm
BE in subjects with lesser gastric acidity, such as around pH 6.8. This rule
does not apply to enteric-coated products. If the use of the drug is limited to
a special population and the test and reference products show a significant
difference in dissolution even under one of the conditions of the dissolution
test, the in vivo test should be performed using subjects from the target
population.
When it is unfavorable to use healthy subjects because of potent pharmaco-
logical action or adverse effects, patients should be employed. If the clearance of
the drug differs to a large extent amongst subjects due to genetic polymorphism,
subjects with higher clearance should be employed.
Japan 171
172 Riku
Washout Periods
Washout periods in cross-over studies between administration of test and refer-
ence products should usually be more than five times the elimination half-life of
the parent compound or active metabolites.
Parameters to be Assessed
When blood samples are used, AUCt and Cmax should be subjected to the BE
assessment used in single-dose studies.
Logarithmic Transformation
The pharmacokinetic parameters, Cmax and AUCt for immediate-release dosage
forms and Cmax and AUC for oral controlled-release products should be statis-
tically analyzed after logarithmic transformation.
Statistical Analysis
The 90% confidence interval or two one-sided t tests with a significance level of
5% should be used. When an add-on subject study is performed and there are no
fundamental differences between the two studies in formulation, design, assay
and subjects, data from the initial and add-on subject studies can be pooled and
statistically analyzed.
It is interesting to note that in Japan, even though the 90% confidence
interval may be outside the 0.8 to 1.25 acceptance range, test products may be
accepted as bioequivalent, provided the following three conditions have been
satisfied:
(1) The total sample size of the initial BE study is not less than 20 (n = 10/group)
or pooled sample size of the initial and add-on subject studies is not less
than 30.
(2) The differences in average logarithmic values of AUC and Cmax between two
products are between log (0.90) to log (1.11).
(3) Dissolution behavior of test and reference products is evaluated to be similar
under all dissolution testing conditions.
Additional metrics such as AUC∞ , tmax , MRT, kel , etc., should also be
subjected to statistical assessment, and if a significant difference is detected in
these reference or secondary parameters between reference and test products, an
explanation must be given to justify that such a difference(s) is/are not consid-
ered to affect the clinical outcomes.
Dissolution Tests
Dissolution tests should be performed, using suitably validated dissolution test
methods and analytical assay.
SPH SPH
IHBK055-07 IHBK055-Kanfer January 6, 2010 20:18 Char Count=
Japan 173
Testing Time
Dissolution testing should generally be performed for two hours in pH 1.2
medium and six hours in other test fluids. However, testing can be terminated
whenever the average dissolution rate of the reference product reaches 85%.
Testing Conditions
Apparatus: Paddle apparatus specified in the JP (12).
Test solutions
(a) Products containing acidic drugs
(i) Test at 50 rpm in the following three types of dissolution media: pH 1.2,
5.5 to 6.5, 6.8 to 7.5, and also in water as well, that is, a single medium
between 5.5 and 6.5, and one between 6.8 and 7.5.
(ii) Test at 100 rpm in either one of the following three types of dissolution
media: pH 1.2, 5.5 to 6.5, 6.8 to 7.5
(b) Products containing neutral or basic drugs, and coated products
(i) Test at 50 rpm in the following three types of dissolution media: pH
1.2, 3.0 to 5.0, 6.8, and also in water as well in each of these media.
(ii) Test at 100 rpm in either one of the following three types of dissolution
media: pH 1.2, 3.0 to 5.0, 6.8
(c) Products containing low-solubility drugs
(i) Test at 50 rpm in the following dissolution media: (1) pH 1.2, (2) pH
4.0, (3) pH 6.8, (4) Water, (5) pH 1.2 + polysorbate 80, (6) pH 4.0 +
polysorbate 80, (7) pH 6.8 + polysorbate 80 and at 100 rpm in either
one of (5) or (6) or (7) above.
(d) Enteric-coated products
(i) Test at 50 rpm in the following dissolution media: (1) pH 1.2,
(2) pH 6.0, (3) pH 6.8, and at 100 rpm in (2) above.
Acceptance Criteria
The average dissolution from the reference product reaches 85% or greater within
15 minutes: the average dissolution from the test product also reaches 85% or
greater within 15 minutes or does not deviate by more than 15% from that of the
reference product at 15 minute.
The average dissolution from the reference product reaches 85% or greater
between 15 and 30 minutes: the average dissolution from the test product does
not deviate by more than 15% from that of the reference product at two time
SPH SPH
IHBK055-07 IHBK055-Kanfer January 6, 2010 20:18 Char Count=
174 Riku
points when the average dissolution from the reference product is around 60%
and 85%.
The average dissolution from the reference product does not reach 85% or
greater within 15 minutes: the criteria are specified in details in the guideline
(10).
PHARMACODYNAMIC STUDIES
These studies are performed to establish the equivalence of products using phar-
macological activity in humans as an assessment index and may be applied to
drug products that do not produce measurable concentrations of the parent drug
or active metabolite in blood or urine or whose BA does not reflect therapeutic
effectiveness.
CLINICAL STUDIES
These studies are performed to establish the therapeutic equivalence of drugs
using clinical endpoints as assessment index. If BE studies and pharmacody-
namic studies are neither possible nor inappropriate, this type of study can be
used.
Test Methods
Bioequivalence studies should be performed using single-dose studies in both
the fasting and fed states. In fed studies, a high-fat diet of 900 kcal or more con-
taining 35% or more lipid content should be used. The meal should be eaten
within 20 minutes, and drugs administered within 10 minutes thereafter.
When the possibility of severe adverse events may occur after dosing in
the fasting state, the fasting dose studies can be replaced with postprandial dose
studies with the low-fat meal employed in the study for immediate-release oral
dosage forms and enteric-coated products (see “Single- Versus Multiple-Dose
Studies” section)
Dissolution Tests
Testing Time
Dissolution testing should generally be performed over a 24-hour period but
may be terminated after two hour in pH 1.2 medium and whenever the average
dissolution rate of the reference product reaches 85%.
SPH SPH
IHBK055-07 IHBK055-Kanfer January 6, 2010 20:18 Char Count=
Japan 175
Testing Conditions
The test should be carried out under the following conditions using the paddle
apparatus and either the rotating basket or disintegration testing apparatus.
Paddle apparatus:
Test at 50 rpm using the following dissolution media: pH1.2, (2) pH 3.0–5.0,
(3) pH 6.8–7.5, (4) water, (5) 1% polysorbate 80 added to pH 6.8–7.5 medium and
at 100 rpm and 200 rpm in dissolution medium of pH 6.8–7.5.
Acceptance criteria for similarity and equivalence of dissolution behavior
If the results meet any one of the following criteria under all testing con-
ditions, the test products are judged to be similar or equivalent to the reference
product when the average dissolution from the reference product reaches 80%
or greater at the time specified under at least one testing condition (see “Similar-
ity of Dissolution Profiles” section). If the f 2 function is used for evaluation, the
judgment is based on the Attachment in the Guideline.
Acceptance Criteria
(1) When the average dissolution from the reference product reaches 80% or
greater at the time specified and the average dissolution from the test prod-
uct does not deviate by more than 15% (in this case, judged as similar) or
10% (in this case, judged as equivalent) from those of the reference product
at three time points when the average dissolution from the reference product
is around 30%, 50%, and 80%.
(2) When the average dissolution from the reference product reaches 50% or
greater but does not reach 80% at the time specified and the average disso-
lution from the test product does not deviate by more than 12% (in this case,
judged as similar) or 8% (in this case, judged as equivalent) from those of
the reference product at the appropriate time point when the average disso-
lution from the reference product is half the average dissolution and at the
time specified.
(3) When the average dissolution from the reference product does not reach
50% at the time specified and the dissolution from the test product does not
deviate by more than 9% (in this case, judged as similar) or 6% (in this case,
judged as equivalent) from those of the reference product at the appropriate
time point when the average dissolution from the reference product is half
the dissolution and at the time specified.
176 Riku
Pharmacokinetic test
Clinical trial
In vitro efficacy test
Animal test
The most suitable test from those already mentioned should be selected by
considering the characteristics of the topical product. If other alternative appro-
priate tests are available, they may be employed.
The testing procedure, analytical method, stability of drugs during storage
and analysis, etc., should be validated. Detailed standard operating procedures
(SOPs) should be prepared for each test, involving the application and removal
of products, recovery of the sample, measurements of pharmacological response,
skin stripping procedures, and analytical method, because the testing procedures
for topical products are generally complicated.
Dermatopharmacokinetics Test
This test is used to assess BE by comparing the amount of active ingredient from
the test and reference products penetrating into the stratum corneum. Topical
drugs are generally distributed into the stratum corneum and reach the epider-
mal cells. Thus, BA in the skin can be estimated by measuring the amount of the
drug in the stratum corneum by means of skin stripping using adhesive tapes. This
method is applicable to topical drugs whose site of action is the stratum corneum
itself or deeper.
Parameters for BE assessment include drug recovery amount or the mean
drug concentration at a steady state in the stratum corneum. The parameters
should be logarithmically transformed and the 90% confidence interval of the
difference in mean values of parameters between reference and test products
should be calculated by a parametric method.
Pharmacological Test
This test is used to assess BE by using the pharmacological response of the topical
product, which correlates with clinical efficacy or BA. Topically applied corticos-
teroids produce a vasoconstrictor effect depending on the drug uptake into the
skin that results in skin blanching. This pharmacological response correlates with
clinical efficacy of topical corticosteroids and can be used as a measure to assess
the BE of topical corticosteroid products. The AUEC (area under the effect curve)
values are used to assess BE of such products. When an instrumental method
is used such as a, chromameter, to measure the degree of skin blanching, the
AUEC values should not be log-transformed. When a visual assay is employed
to measure skin blanching, the 90% confidence interval of the difference in mean
AUEC values between test and reference products is calculated by a nonpara-
metric method or parametric method after the logarithmic transformation of the
AUEC values (14).
Japan 177
this test may be useful if precise measurements can be made. The amount dis-
tributed into the skin following application of the product is used as a measure
of BE. In principle, the data should be logarithmically transformed, and the 90%
confidence interval of the difference of the mean parameter values between test
and reference products is calculated by a parametric method.
Pharmacokinetic Test
This test is used to assess BE by using pharmacokinetic parameters from blood
concentration–time curves after product application. Pivotal tests should be per-
formed according to the 1997 Guideline (4). AUC or Css (steady-state drug con-
centration) is used as the parameter to assess BE. In principle, the data should be
logarithmically transformed, and the 90% confidence interval confidence of the
difference in mean parameter values between test and reference products should
be calculated by a parametric method.
Animal Tests
This test is used to assess BE by using a pharmacological response produced
on the skin. The test may be applicable for topical drugs such as bactericides,
disinfectants, antiseptics, hemostatics, and wound repair agents whose active site
is the surface of the skin and which are not intended to penetrate the stratum
corneum. The appropriate acceptable range for BE should be established for each
drug to assess BE.
178 Riku
Different Strengths
Bioequivalence studies for different strengths of solid oral dosage forms, such
as additional strength(s) of the same dosage form, for example adding a
5-mg tablet to a series of approved tablets of different strengths, should follow
the “Guideline for Bioequivalence Studies for Different Strengths of Oral Solid
Dosage Forms 2000” (5) and the 2006 revision (10), which applies to products
that contain the same active ingredient, in the same dosage form and used in the
same dosage regimen as the product already approved but differing in strength.
Reference Product
The reference product is the original innovator product that has previously been
approved in Japan on the basis of therapeutic efficacy and safety data established
by clinical trials, and is selected from three batches of the innovator product on
the domestic market. Dissolution tests are generally performed on three lots of
the innovator available on the domestic market to select the appropriate batch for
use as the reference product in the biostudy. Among the three lots, the one which
shows intermediate dissolution patterns between the lots should be selected as
the reference product. It should be noted that the reference product need not
always be the innovator product but can sometimes also be an approved generic
product.
For example, where a range of innovator products, such as say, a 10-mg
and 20-mg tablet are available on the market in Japan, and only a 10-mg generic
product has previously been approved on the basis of a BE study. Should the
same applicant wish to market a 20-mg generic tablet, either the 20-mg innovator
product or the 10 mg previously approved generic can be used as a reference
product.
Japan 179
180 Riku
Japan 181
REFERENCES
1. Ministry of Health, Labor and welfare. Enforcement of the partial amendments to
Japanese Pharmaceutical Affairs Law. Yakuhatsu Notification No.421, March 27, 2007.
2. Japanese Pharmaceutical Affairs Law. Article 14, Paragraph 3, June, 1996.
3. Annette Cunningham. Bolar Provision: A global history and the future for Europe.
http://www.genericsweb.com/index.php?object id = 238.
4. Ministry of Health, Labor and Welfare. Guideline for Bioequivalence Stud-
ies of Generic Products, 1997. http://www.nihs.go.jp/drug/be-guide(e)/Generic/
be97E.html.
5. Ministry of Health, Labor and Welfare. Guideline for Bioequivalence Studies for Dif-
ferent Strengths of Oral Solid Dosage Forms, 2000. http://www.nihs.go.jp/drug/be-
guide(e)/strength/strength.html.
6. Ministry of Health, Labor and Welfare. Guideline for Bioequivalence Studies for
Additional Dosage Forms of Oral Solid Dosage Forms, 2001.
7. Ministry of Health, Labor and Welfare. Guideline for Bioequivalence Studies of
Generic Products for Topical Dermal Application, 2003. http://www.nihs.go.jp/
drug/be-guide(e)/Topical BE-E.pdf.
8. Ministry of Health, Labor and Welfare. Guideline for Formulation Changes of Oral
Solid Dosage Forms, 2000. http://www.nihs.go.jp/drug/be-guide(e)/form/form-
change.PDF.
9. Ministry of Health, Labor and Welfare. Draft Guideline for Bioequivalence Stud-
ies for Changes in Manufacturing of Oral Solid Dosage Forms: Immediate-release
(Conventional) and Enteric Coated Products, 2002.
10. Ministry of Health, Labor and Welfare. Revision of Guideline for Bioequivalence
Studies of Generic Products, 2006.
11. WHO Expert Committee. WHO Technical Report Series No. 863: Multisource
(generic) pharmaceutical products: WHO guidelines on registration requirements
to establish interchangeability, 1996. http://apps.who.int/medicinedocs/es/d/
Js5516e/19.html.
12. Ministry of Health, Labor and Welfare. Japanese Pharmacopoeia 15th 2006. English
ed. 116. http://jpdb.nihs.go.jp/jp15e/JP15.pdf.
13. Ministry of Health, Labor and Welfare. Q & A on Revision of Guideline for Bioequiv-
alence Studies of Generic Products, Shinsakanrika, Administrative Communication,
November 24, 2006.
14. Hauschke D, Steinijans VW, Diletti E. A distribution—Free procedure for the statisti-
cal analysis of bioequivalence studies. J Clin Pharmacol Ther Toxicol 1990; 28(2):72–78.
15. Ministry of Health, Labour and Welfare. Q & A on Guideline for Bioequivalence Stud-
ies for Additional Dosage Forms of Oral Solid Dosage Forms, 2001. Shinsakanrika,
Administrative Communication, May 31, 2001.
16. Center for Drug Evaluation and Research. Guidance for Industry, Immediate Release
Solid Oral Dosage Forms—Scale-Up and Postapproval Changes: Chemistry, Man-
ufacturing, and Controls, In Vitro Dissolution Testing, and In Vivo Bioequivalence
Documentation. Center for Drug Evaluation and Research (CDER), November
1995, CMC5. Federal Register, vol. 60, no. 230. http://www.fda.gov/downloads/
Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070636.pdf
17. Nobuo Aoyagi. Japanese guidance on bioavailability and bioequivalence. Eur J Drug
Metab Pharmacokinet 2000; 27(1):28–31.
18. Ministry of Health, Labor and Welfare. Q & A on Guideline for Bioequivalence
Studies of Generic Products, 1997, Shinsakanrika, Administrative Communication,
October 30, 1998.
19. Kiyoto Nakai, Masahiko Fujita, and Hiroyuki Ogata. International harmonization
of bioequivalence studies and issues shared in common. Yakugaku Zasshi 2000;
120(11):1193–1200.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
8 South Africa
Isadore Kanfer and Roderick B. Walker
Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa
Michael F. Skinner
Biopharmaceutics Research Institute, Rhodes University, Grahamstown,
South Africa
INTRODUCTION
The Medicines Control Council (MCC) in South Africa is a statutory body that
was established in terms of the Medicines and Related Substances Control Act
(MRSCA), 101 of 1965, to oversee the regulation of medicines in South Africa.
Applicants are required to submit evidence of quality, safety, and efficacy for
new drugs and medicinal products as well as for the registration of generic (mul-
tisource) medicinal products. In the latter instance, bioequivalence (BE) data can
be used as a surrogate measure of safety and efficacy. To facilitate the registration
process for generic medicines, guidelines have been prepared to serve as a rec-
ommendation to applicants wishing to submit data in support of the registration
of such medicines (1–4).
The mandate of the South African MCC is to safeguard and protect the pub-
lic through ensuring that all medicines that are sold and used in South Africa
are safe, therapeutically effective, and consistently meet acceptable standards
of quality. In this respect, all submissions must provide the necessary data for
quality, safety, and efficacy to register an interchangeable multisource (generic)
pharmaceutical product (medicine) and thereby infer that it is therapeutically
equivalent.
Several important definitions and specific terms have been described in
the relevant Act (5) as well as in the associated guidelines. For the most part,
these considerations parallel the requirements for multisource interchangeabil-
ity as defined within the United States and the European Union (EU). However,
a notable exception is that the reference (comparator) product used in the BE
study need not be the domestic innovator product. This provision permits BE
data generated between the test (generic) product and a foreign reference prod-
uct to be submitted as proof of safety and efficacy. The implication here is that
a BE study undertaken with the primary objective of gaining market access in a
foreign country is simply “piggy-backed” in the dossier for South African reg-
istration as a secondary consideration. This has resulted in the development of
a market for the sale of dossiers where the same BE data are used by several
generic drug companies to gain access to the South African market without the
need to undertake the necessary studies to establish interchangeability between
that generic and the innovator products being sold in South Africa.
182
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
Historical Background
Proof of safety and efficacy of generic medicines have in the past been based
upon requirements described in “official” notices or circulars issued by the MCC.
In fact, registration requirements for generic medicines, particularly with respect
to safety and efficacy, have not been well defined until fairly recently. For exam-
ple, in some instances BE testing was mentioned as a requirement whereas in
others this requirement has been optional depending on the interpretation of the
MCCs issued notices and/or circulars. In many instances, only in vitro dissolu-
tion testing was required on the basis of Circular 14/95 that was first issued in
the early 1990s and subsequently updated in 1995 (6).
Part 2.2.1 of Circular14/95 (6) (see Appendix 1) provided for the use of
comparative dissolution studies of the test and reference products as a proof of
efficacy. The dissolution requirements were contingent on there being a United
States Pharmacopeia (USP) monograph for the active ingredient, which included
dissolution requirements and provided that the active was not included in a com-
piled list, which contained 96 drugs and drug combinations listed in alphabetic
order (Appendix 1). No reasons were given why those specific compounds were
on the list. The assay results of both the reference and test products were required
as well as content uniformity test results and dissolution testing in three media
where required as follows:
Dissolution testing was required to be carried out according to the USP dis-
solution requirements specified in the monograph using at least six units of the
product, the apparatus, medium volume, and rotation speed. For the other two
media, a further six units of both the test and reference products were required
to be tested using the specified USP dissolution apparatus, medium volume, and
rotation speed specified in the monograph.
All dissolution testing was required to be multipoint studies and the results
presented in a tabulated and graphical form. However, no indication was given
regarding acceptance criteria for the declaration of BE. In the case where the
active is insoluble in the other two media, a motivation could be submitted to
the MCC for the omission of further testing in the other two media.
Proof of efficacy of vitamins or vitamins and mineral combinations and also
for phenolphthalein and sucralfate products was accepted on the basis of disin-
tegration testing where the disintegration test needed to be carried out according
to the USP disintegration test for nutritional supplements for the vitamins or the
general disintegration test for the other substances.
The requirements for proof of efficacy for the following types of products
are listed here:
1) For all products with an antacid or acid neutralizing claim, the acid neutral-
izing capacity test included in the USP was required.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
This guideline (i.e., Circular 14/95) made provision for the use of any other
method, which an applicant wished to submit, provided the rationale for the
particular method was included.
It is interesting to note that there are still products on the South African
market, which were registered according to the requirements described in Cir-
cular 14/95 (6). A matter of concern here is that in many cases the innovator
product used for the comparison may no longer be on the market and therefore
a generic product that is the market leader and as such required to be used as
the reference product in BE testing may well be a product that has never been
assessed for safety and efficacy in a BE study.
It was only in the early 2000s that the requirements for the registration and
market approval of generic medicines were published as official guidelines. Dur-
ing 2002, legislation that required that multisource (generic) medicines registered
in South Africa must be offered to all patients when a physician prescribes an
innovator product was introduced.
In terms of the Medicines and Related Substances Control Act, 1965 (Act
No. 101 of 1965) as amended by Act No. 90 of 1997 (5) and Act 59 of 2002 (7),
provision is made for generic substitution in Section 22F.
“Subject to subsections (2), (3), and (4) of the Act, a pharmacist or a person
licensed in terms of Section 22C shall:
a) inform all members of the public who visit the pharmacy or any other place where
dispensing takes place, as the case may be, with a prescription for dispensing, of
the benefits of the substitution for a branded medicine by an interchangeable multi-
source medicine; and
b) dispense an interchangeable multi-source medicine prescribed by a medical practi-
tioner, dentist, practitioner, nurse or other person registered under the Health Pro-
fessions Act, 1974, unless expressly forbidden by the patient to do so.”
Furthermore, subsection (4) states that “A pharmacist shall not sell an inter-
changeable multi-source medicine
r if the person prescribing the medicine has written in his or her own hand on the
prescription the words ‘no substitution’ next to the item prescribed;
r if the retail price of the interchangeable multi-source medicine is higher than that of
the prescribed medicine; or
r where the product has been declared not substitutable by the council.” (4)
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
Study Design
This is described under Section 3.1 of the Biostudies guidelines (2), which is
introduced with the statement “The study should be designed in such a way
that the formulation effect can be distinguished from other effects. If the num-
ber of formulations to be compared is two, a balanced two-period, two-sequence
crossover design is considered to be the design of choice.” Other designs such
as well-established parallel designs for very long half-life substances may also
be considered provided the study design and the statistical analyses are scientif-
ically sound.
Generally, single-dose studies are recommended but provision is made to
also allow steady-state studies provided such a study can be justified.
content are more likely to affect the gastrointestinal (GI) physiology and thereby
result in a larger effect on BA, the use of high-calorie and high-fat meals during
food-effect studies is recommended.
Number of Subjects
The number of subjects should be justified on the basis of providing at least
80% power of meeting the acceptance criteria. The minimum number of sub-
jects should not be less than 12. If 12 subjects do not provide 80% power, more
subjects should be included.
A minimum of 20 subjects is required for modified-release oral dosage
forms.
Add-On Subjects
If the BE study was performed with the appropriate size but BE cannot be
demonstrated because of a result of a larger than expected random variation or
a relative difference, an add-on subject study can be performed using not more
than the number of subjects in the initial study. Combining is acceptable only
in the case when the same protocol was used and preparations from the same
batches were used.
Provision for an add-on study must be included a priori in the protocol and
carried out strictly according to the study protocol and standard operating pro-
cedures (SOPs), and must be given appropriate statistical treatment, including
consideration of consumer risk.
Subject Selection
BE studies should normally be performed with healthy volunteers. The inclu-
sion/exclusion criteria should be clearly stated in the protocol.
In general, the following subject characteristics are required:
i) Sex—Both male and female subjects can be included but the risk to women
of childbearing potential should be considered on an individual basis.
ii) Age—Subjects should be between 18 and 55 years.
iii) Body mass—Subjects should have a body mass within the normal range
according to accepted normal values for the body mass index (BMI = mass
in kg divided by height in meters squared, i.e., kg/m2 ), or within 15% of the
ideal body mass, or any other recognized reference.
iv) Informed consent—All subjects participating in the study should be capa-
ble of giving informed consent.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
Inclusion of Patients
If the API under investigation is known to produce adverse effects and the phar-
macological effects or risks are considered unacceptable for healthy volunteers, it
may be necessary to use patients instead, under suitable precautions and super-
vision. In this case the applicant should justify the use of patients instead of
healthy volunteers.
Genetic Phenotyping
Phenotyping and/or genotyping of subjects may be considered in crossover
studies (e.g., BE, dose proportionality, food interaction studies) for safety or
pharmacokinetic reasons. If an API is known to be subject to major genetic poly-
morphism, studies could be performed in cohorts of subjects of known pheno-
type or genotype for the polymorphism in question.
Dosing
The time of day for ingestion of doses should be specified.
Concomitant Medication
Subjects should not take other medicines for a suitable period prior to, and dur-
ing, the study and should abstain from food and drinks, which may interact with
circulatory, GI, liver, or renal function (e.g., alcoholic or xanthine-containing bev-
erages or certain fruit juices).
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
Sampling Frequency
The sampling schedule should be planned to provide an adequate estimation
of Cmax and to cover the plasma drug concentration time curve long enough to
provide a reliable estimate of the extent of absorption. This is generally achieved
if the AUC derived from measurements is at least 80% of the AUC extrapolated
to infinity.
If a reliable estimate of terminal half-life is necessary, it should be obtained
by collecting at least three to four samples above the limit of quantitation (LOQ)
during the terminal log-linear phase.
For long half-life drugs/APIs (>24 hours), the study should cover a mini-
mum of 72 hours, unless 80% is recovered before 72 hours. The guidance states
that “for moieties demonstrating high inter-subject variability in distribution and
clearance the use of AUC truncation warrants caution. In these circumstances
sampling periods beyond 72 hours may be required.”
To allow accurate estimation of relevant parameters, sampling points
should be chosen such that the plasma concentration versus time profiles can
be adequately defined.
Blood Sampling
a) The blood sampling frequency and duration should be sufficient to account
for at least 80% of the known AUC to infinity (AUC∞ ), usually approxi-
mately three terminal half-lives of the drug/API.
b) For most drugs/APIs 12 to 18 samples including a predose sample should be
collected per subject per dose.
c) Sample collection should be spaced such that the maximum concentrations
of drug/API in blood (Cmax ) and the terminal elimination rate constant (Kel )
can be estimated.
d) At least three to four samples above the LOQ should be obtained during the
terminal log-linear phase to estimate Kel by linear regression analysis.
e) The actual clock time when samples are collected, as well as the elapsed time
relative to drug/API administration should be recorded.
Urine Sampling
a) Volumes of each sample should be measured immediately after collection
and included in the report.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
b) Urine should be collected over an extended period and generally no less than
seven times the terminal elimination half-life, so that the amount excreted to
infinity (Ae∞ ) can be estimated.
c) Sufficient samples should be obtained to permit an estimate of the rate and
extent of renal excretion. For a 24-hour study, sampling times of 0 to 2, 2 to 4,
4 to 8, 8 to 12, and 12 to 24 hours postdose are usually appropriate.
d) The actual clock time when samples are collected, as well as the elapsed time
relative to API administration should be recorded.
CHARACTERISTICS TO BE INVESTIGATED
Moieties To Be Measured
Parent or Metabolite(s)
The evaluation of BA and BE should, in general, be based on measured concen-
trations of the parent compound (i.e., the active). The determination of moieties
should be measured in biological fluids to take into account both concentration
and activity.
The guideline recommends that for a BA study, both the parent active and
its major active metabolites should be measured, if analytically feasible. This is
necessary to assess both the concentration and the relative contribution of both
the active parent and its major active metabolite(s) in the biological fluid to the
clinical safety and/or efficacy of the active component(s).
However, for BE determinations, measurement of only the active parent
released from the dosage form, rather than any metabolite(s), is generally recom-
mended. The rationale for this recommendation is that the concentration–time
profile of the active parent is more sensitive to changes in formulation perfor-
mance than a metabolite, which is more reflective of metabolite formation, dis-
tribution, and elimination.
Notwithstanding, it is important to state a priori in the study protocol
which chemical entities (pro-drug, API, metabolite) will be analyzed in the
samples.
In some situations, however, measurements of an active or inactive metabo-
lite may be necessary instead of the parent compound. Instances where this may
be necessary are as follows:
PHARMACOKINETIC PARAMETERS
a) AUCt , AUC∞ , Cmax , tmax for plasma concentration versus time profiles.
b) AUC∞ , Cmax , Cmin , fluctuation (% PTF), and swing (% swing) for studies
conducted at steady state.
c) Any other justifiable characteristics as referred to in Appendix I.
d) The method of estimating AUC values should be specified.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
PHARMACODYNAMIC STUDIES
If pharmacodynamic parameters/effects are used as BE criteria, the applicant
must submit justification for their use. In addition
BIOANALYSIS
Bioanalysis of all analytes must be conducted according to good laboratory prac-
tice (GLP) and cGMP. All analytical methods used should be fully validated and
documented. The following characteristics of the assay need to be addressed:
STUDY PRODUCTS
Reference Products
Reference to another guideline entitled Pharmaceutical and Analytical (P&A)
Guideline (1) is made under this section. The requirements relating to pharma-
ceutical and analytical information are provided in this guideline, including ele-
ments of pharmaceutical and biological availability in Part 2A—Basis for Regis-
tration and Overview of Application. Although there is a Part 2C section, Quality
Overall Summary (QOS), PART 2B is conspicuously absent. Perusal of the P&A
guideline reveals the following:
Under Section 2.1.3 headed as Study Products and subsection (b) headed
Reference Products (comparators) (see also Biostudies and Dissolution Guidelines), a
statement is made that
It is also stated that the reference product may contain a different chemi-
cal form from that of the proposed generic product (pharmaceutical alternative)
provided it is confirmed that the safety/efficacy profile is not altered. Further-
more, if well known (e.g., hydrochloride, maleate, nitrate, stearate), reference to
a pharmacopoeia accepted by the council (i.e., MCC) may be acceptable. This is
in direct conflict with the provisions of the Medicines and Related Substances
Act as amended (5), wherein it is unambiguously indicated that such compar-
isons do not qualify for assessment as interchangeable medicines, since they are
not pharmaceutically equivalent and thus cannot be declared bioequivalent In
order to make provision for a pharmaceutical alternative, the Act would need to
be amended accordingly to incorporate the new definition, since the guideline is
enabled by the Act and not vice versa.
Product strengths not available in South Africa may be applied for and/or
used in biostudies provided that the dose range is approved/registered in South
Africa.
The reference product should be an innovator product registered by the
MCC and should preferably be procured in South Africa. An exception is an “old
medicine” that may be used as a reference product when no other such product
has been registered provided that it is available on the South African market.
If more than one such product is available the market leader should be used as
the reference (e.g., IMS database) and the applicant has to submit evidence to
substantiate the market leadership claim.
Quite extraordinarily, several options for the selection of the reference
product are given and listed in order of preference:
(iii) A product from the latest edition of the WHO International compara-
tor products for equivalent assessment of interchangeable multisource
(generic) products QAS/05.143.a or;
(iv) In the case that no innovator product can be identified—within the context
of (i) to (iii) above, the choice of the reference must be made carefully and
must be comprehensively justified by the applicant.
Although it is noted that “A product that has been approved based on com-
parison with a non-domestic reference product may or may not be interchange-
able with currently marketed domestic products,” the significance and implica-
tion of the foregoing statement is unclear? The guidelines state that in all cases,
the choice of reference product should be justified by the applicant and the coun-
try of origin of the reference product should be reported together with lot num-
ber and expiry date. Recently, an amended guidelineb has included the following
requirements for applicants using a foreign reference product.
(i) The name and address of the manufacturing site where the foreign reference product
is manufactured.
(ii) The qualitative formulation of the foreign reference product.
(iii) Copies of the immediate container label as well as the carton or outer container
label of the foreign reference product.
(iv) For modified release, evidence of the mechanism of modified release of the foreign
reference product.
(v) The method of manufacture of the foreign reference product if claimed by the appli-
cant to be the same.
(vi) Procurement information of the foreign reference product
r Copy of licensing agreement/s
r Distribution arrangements/agreement/s
r Copy of purchase invoice (to reflect date and place of purchase)
It is interesting to note that no other specific conditions for the use of a for-
eign (non-domestic) reference product are mentioned until Section 5.1.2 of the
Biostudies guideline (2). It is therefore highly likely that different generic prod-
ucts may well be or have been approved following comparison to different non-
domestic reference products without comparisons having been made between
different nondomestic reference products (the implications for generic substitu-
tion and therapy, in general are obvious). Section 4.2 of the Dissolution guidelines
and “Foreign Reference Products” section of this chapter describe the provisions
made for the use of a foreign reference product and the comparative dissolution
testing required to determine whether such a foreign reference product complies
with the specified requirements for use in BE studies.
The choice of reference products for combination products makes reference
to the Biostudies (2) and Dissolution guidelines (3) and states that such products
a http://www.who.int/medicines/services/expertcommittees/pharmprep/QAS05 143
Comparator Rev%201.pdf. Accessed June 10, 2009.
b Pharmaceutical and Analytical: Medicines Control Council, Department of Health, 2.02 P&A
Apr09 v3.doc. http://www.mccza.com/documents/2.02 P&A Apr09 v3.doc. Accessed June
11, 2009.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
Retention Samples
A sufficient number of retention samples of both test and reference products used
in the BE study should be kept for one year in excess of the accepted shelf life, or
two years after completion of the trial or until approval, whichever is longer, to
allow re-testing if required by the MCC.
DATA ANALYSIS
Statistical Analysis
The guideline states that “The statistical method for testing relative bioavailabil-
ity (i.e. average bioequivalence) is based upon the 90% confidence interval for the
ratio of the population means (Test/Reference) for the parameters under consid-
eration. Pharmacokinetic parameters derived from measures of concentration,
e.g. AUCt , AUC∞ and Cmax should be analysed using ANOVA. Data for these
parameters should be transformed prior to analysis using a logarithmic transfor-
mation.”
If appropriate to the evaluation, the analysis technique for tmax should be
nonparametric and should be applied to untransformed data.
In addition to the appropriate 90% confidence intervals, summary statistics
such as geometric and arithmetic means, SD and% RSD, as well as ranges for
pharmacokinetic parameters (minimum and maximum), should be provided. A
disk with raw data formatted appropriately for evaluation where the formatting
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
STUDY REPORT
Complete documentation is required to be submitted including the protocol, con-
duct, and evaluation and evidence of compliance with GCP, GLP, and cGMP.
Clinical Report
The following information must be included in the clinical section of the BE study
report:
a) A statement indicating the independence of the ethics committee.
b) Documented proof of ethical approval of the study.
c) A complete list of the members of the ethics committee, their qualifications,
and affiliations.
d) Names and affiliations of the all investigator(s), the site of the study and the
period of its execution.
e) The names and batch numbers of the products being tested.
f) The name and addresses of the applicants of both the reference and the test
products.
g) Expiry date of the reference product and the date of manufacture of the test
product used in the study.
h) Assay and dissolution profiles for test and reference products.
i) Certificate of analysis of the API used in the test product biobatch.
j) A summary of adverse events, which should be accompanied by a discussion
on the influence of these events on the outcome of the study.
k) A summary of protocol deviations (sampling and nonsampling), which
should be accompanied by a discussion on the influence of these adverse
events on the outcome of the study.
l) Subjects who dropout or are withdrawn from the study should be identified
and their withdrawal fully documented and accounted for.
Analytical Report
The following must be included in the analytical section of the BE report:
a) Validation report.
b) All individual subject concentration data.
c) Calibration data, that is, raw data and back-calculated concentrations for
standards, as well as calibration curve parameters, for the entire study.
d) Quality control samples for the entire study.
e) Chromatograms from analytical runs for 20% of all subjects (or for a min-
imum of four subjects, whichever is the greater) including chromatograms
for the associated standards and QC samples.
f) A summary of protocol deviations, which should be accompanied by a dis-
cussion on the influence of these deviations on the outcome of the study.
Protocol deviations should be justified.
These data should be submitted in hard copy and also formatted elec-
tronically in a format compatible for processing by SAS software. Individual
subject data should be in rows and arranged in columns, which reflect the
subject number, phase number, sequence, formulation, and sample concen-
tration versus time data.
b) The method(s) and programmes used to derive the pharmacokinetic param-
eters from the raw data.
c) A detailed ANOVA and/or nonparametric analysis, the point estimates and
corresponding confidence intervals for each parameter of interest.
d) Tabulated summaries of pharmacokinetic and statistical data.
e) The statistical report should contain sufficient detail to enable the statistical
analysis to be repeated, for example, individual demographic data, random-
ization scheme, individual subject concentration versus time data, values of
pharmacokinetic parameters for each subject, descriptive statistics of phar-
macokinetic parameters for each formulation and period.
Quality Assurance
The study report should contain a signed quality assurance (QA) statement con-
firming release of the document. The applicant should indicate whether the
site(s) (clinical and analytical) where the study was performed was subjected
to a prestudy audit to ascertain its/their status of GCP and GLP and/or cGMP
conditions. All audit certificates should clearly indicate the date of audit and
the name(s), address(es) and qualifications of the auditor(s) and, in addition, an
independent monitor’s statement must be included.
Mention is made in the guideline that the applicant must demonstrate
that the excipients in the pharmaceutically equivalent product are essentially
the same and in comparable concentrations as those in the reference product.
Interestingly, no mention is made of the use of a pharmaceutical alternative as
provided in the guideline. In the event that this information about the reference
product cannot be provided by the applicant, it is incumbent upon the appli-
cant to perform in vivo or in vitro studies to demonstrate that the differences in
excipients do not affect product performance.
Suspensions
BE for a suspension should be treated in the same way as for immediate-release
solid oral dosage forms.
Modified-Release Products
BE studies (single dose) are required for these dosage forms, which include
delayed-release products and extended- (controlled-)release products (as defined
in the P&A guideline). Fasted as well as fed studies are necessary and multiple-
dose studies are generally not recommended.
Parenteral Solutions
The applicant must demonstrate that the excipients in the pharmaceutically
equivalent (no mention made of pharmaceutical alternative dosage forms?)
product are essentially the same and in comparable concentrations as those in the
reference product. In the event that this information about the reference product
cannot be provided by the applicant, it is incumbent upon the applicant to per-
form in vivo or in vitro studies to demonstrate that the differences in excipients
do not affect product performance. The nature of such studies are however, not
disclosed.
Aqueous Solutions
Aqueous solutions to be administered by parenteral routes (intravenous,
intramuscular, subcutaneous) containing the same active pharmaceutical ingre-
dient(s) in the same molar concentration and the same or similar excipients
in comparable concentrations as the comparator product are considered to be
equivalent without the need for further documentation.
Certain excipients (e.g., buffer, preservative, antioxidant) may be different
provided the change in these excipients is not expected to affect the safety and/or
efficacy of the medicine product.
Other
BE studies are required for all other parenterals and for intramuscular dosage
forms, monitoring is required until at least 80% of the AUC∞ has been covered.
TOPICAL PRODUCTS
The guideline states that “Pharmaceutically equivalent topical products pre-
pared as aqueous solutions containing the same active pharmaceutical ingredi-
ent(s) in the same molar concentration and essentially the same excipients in
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
Local Action
For topical preparations containing corticosteroids intended for application to
the skin and scalp, the human vasoconstrictor test (blanching test) is recom-
mended for BE assessment of such products. Either visual or chromameter data
are acceptable but in each case all data must be validated.
For simple topical solutions with bacteriostatic, bactericidal, antiseptic,
and/or antifungal claims, a biowaiver based on appropriate validated in vitro
test methods, for example, microbial growth inhibition zones, is acceptable.
For all other topical formulations, clinical data (comparative clinical effi-
cacy) are required. Proof of release by membrane diffusion is not acceptable
as proof of efficacy, unless data are presented that show a correlation between
release through a membrane and clinical efficacy.
The guideline also states that whenever systemic exposure resulting from
locally applied/locally acting medicinal products entails a risk of systemic
adverse reactions, systemic exposure should be measured.
Systemic Action
A BE study is always required for other locally applied products with systemic
action, for example, transdermal products.
Gases
Pharmaceutically equivalent gases are considered to be equivalent without the
need for further documentation.
Immediate-Release Tablets
When the pharmaceutical product is the same dosage form but of a different
strength and is proportionally similar in its API and inactive pharmaceutical
ingredients (IPIs), a biowaiver may be acceptable.
Modified-Release Products
Beaded Capsules—Lower Strength
For extended-release beaded capsules where the strength differs only in the num-
ber of beads containing the API, a single-dose, fasting BE study should be carried
out on the highest strength. A biowaiver for the lower strength based on disso-
lution studies can be requested.
Dissolution profiles in support of a biowaiver should be generated for each
strength by using the recommended dissolution test methods described in Sec-
tion 3 of the Dissolution guideline (3).
Dissolution of test and reference products should be conducted in each of
the following three media:
r Acidic media such as 0.1 N HCl
r pH 4.5 buffer
r pH 6.8 buffer
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
If both the test and reference products show more than 85% dissolution
within 15 minutes, the profiles are considered similar (no calculations required).
If not, the f 2 value must be calculated. If f 2 ≥ 50, the profiles are normally
regarded similar such that further in vivo studies are not necessary. Note that
only one measurement should be considered after 85% dissolution of both prod-
ucts has occurred and excluding point zero.
The similarity factor (f 2 ) is a logarithmic reciprocal square-root transforma-
tion of the sum of squared errors, and is a measurement of the similarity in the
percentage (%) dissolution between the two curves, viz.:
f 2 = 50 log{[1 + (1/n) n
(Rt − Tt)2 ]−0.5 .100}
t=1
where n is the number of time points, Rt is the dissolution value of the reference
batch at time t, and Tt is the dissolution value of the test batch at time t.
The dissolution profile of two products, that is, of the test and reference
products (using 12 units each) should be determined and for f 2 calculations, a
minimum of three time points (excluding point zero) must be used, and only one
measurement included after 85% dissolution of both products has occurred.
Generally, f 2 values greater than 50 (50–100) ensure sameness or equiva-
lence of the two curves and, thus, of the performance of the test and reference
products. For curves to be considered similar, f 2 values should be close to 100.
This model-independent method is considered to be most suitable for dis-
solution profile comparisons when three to four or more dissolution time points
are available. The following recommendations should also be considered:
i) The dissolution measurements of the test and reference batches should be
made under exactly the same conditions. The dissolution time points for
both profiles should be the same (e.g., 10, 15, 20, 30, 45, 60 minutes, etc.).
ii) Only one measurement should be considered after 85% dissolution of both
products have occurred.
iii) To allow use of mean data, the percent coefficient of variation (CV) at the
earlier time points (e.g., 15 minutes) should not be more than 20%, and at
other time points should not be more than 10%.
Tablets—Lower Strength
For extended-release tablets when the pharmaceutical product is
a) in the same dosage form but in a different strength,
b) is proportionally similar in its APIs and IPIs, and
c) has the same drug/API release mechanism.
An in vivo BE determination of one or more lower strengths may be waived
on the basis of dissolution testing as previously described. Dissolution profiles
should be generated on all the strengths of the test and the reference products.
When the highest strength (generally, as usually the highest strength is
used unless a lower strength is chosen for reasons of safety) of the multisource
product is bioequivalent to the highest strength or dosee of the reference product,
e Dose included in the dosage range of the MCC-approved package insert of the innovator
product registered in South Africa.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
and other strengths are proportionally similar in formulations and the dissolu-
tion profiles are similar between the dosage strengths, biowaivers can be consid-
ered for the lower/other strengths.
Postregistration/Approval Amendments
The Biostudies guideline comments primarily on registration requirements for
multisource pharmaceutical products, however, it also states that in vitro disso-
lution testing may also be suitable to confirm similarity of product quality and
performance characteristics with minor formulation or manufacturing changes
after approval.
Section 4.3 of the Dissolution guideline describes the types of dissolution
testing required when amendments are made to pharmaceutical products, man-
ufacturing procedures, and other associated processes including change of site.
Three different cases are described, viz.:
a) Case A
Dissolution testing should be conducted as a release test according to the original
submission, or in accordance with compendial requirements, for that product.
b) Case B
Dissolution testing should be conducted as a multipoint test in the applica-
tion/compendial medium at intervals such as 10, 15, 20, 30, 45, 60, and 120 min-
utes, or until an asymptote is reached for the proposed and currently registered
formulation.
c) Case C
Dissolution testing should be conducted as a multipoint test in the three dissolu-
tion media as specified above for the proposed, and currently registered formula-
tions, at intervals such as 10, 15, 20, 30, 45, 60, and 120 minutes, or until either
85% dissolution is obtained, or an asymptote is reached.
a) Type A
In the event that the Type A change made is such that it is unlikely to have
an effect on the quality and performance of a dosage form, Case A dissolu-
tion testing is appropriate.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
b) Type B
In the event that the changes, which were made, could have a significant
impact on the quality and performance of a dosage form, Case B or C disso-
lution testing is appropriate. Profiles of the currently used product and the
proposed product should be proven to be similar, according to the require-
ments as describe in this Guideline.
c) Type C
In the case of changes that are likely to have a significant impact on for-
mulation quality and performance, in vivo bioequivalence testing should
be conducted unless otherwise justified. Case B or Case C dissolution test-
ing may also be required. Biowaivers may also be considered if a proven in
vitro/in vivo correlation (IVIVC) has been established.
CONCLUSIONS
A unique and potentially problematic situation currently exists in South Africa.
Generic substitution has been mandated as a means to ensure that accessibility
to affordable medicines is achieved. Prescribers and dispensers of medicines
must therefore inform patients that prescribed medicines may be available at a
lower price than the innovator/Brand product and recommend accordingly. In
other words, where an approved generic medicine exists, the generic medicine
will be substituted for the innovator/brand product unless the dispenser is
prohibited from doing so by the patient. Clearly, the substitution must be
based on the premise that approved generic medicines have been deemed to
be interchangeable following assessment by the MCC. Of particular concern is
the fact that most generic medicines approved and marketed in South Africa do
not comply with internationally accepted requirements for interchangeability as
many/most have not been assessed by comparison with the innovator/brand
product available on the South African market but rather against a “foreign”
reference product as permitted in the national guidelines. A foreign reference
product, although being supplied by the same innovator/brand company, may
not be the same (identical formulation, manufacture etc.) as the innovator/brand
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
REFERENCES
1. Pharmaceutical and Analytical: Medicines Control Council, Department of
Health, 2.02 P&A, Jun07, v2, 2007. http://www.mccza.com/showdocument
.asp?Cat=17&Desc=Guidelines%20-%20Human%20Medicines. Accessed April 29,
2009.
2. Biostudies: Medicines Control Council, Department of Health, 2.06 Biostudies, Jun 07,
v2, 2007. http://www.mccza.com/showdocument.asp?Cat=17&Desc=Guidelines%
20-%20Human%20Medicines. Accessed April 29, 2009.
3. Dissolution: Medicines Control Council, Department of Health, 2.07 Dissolu-
tion, Jun07, v2, 2007. http://www.mccza.com/showdocument.asp?Cat=17&Desc=
Guidelines%20-%20Human%20Medicines. Accessed April 29, 2009.
4. Generic Substitution: Medicines Control Council, Department of Health, December
2003. http://www.mccza.com/showdocument.asp?Cat=17&Desc=Guidelines%20-
%20Human%20Medicines. Accessed April 29, 2009.
5. Medicines and Related Substances Control Act, 1965 (Act No. 101 of 1965) as amended
by Act No. 90 of 1997 and Act No. 59 of 2002. http://www.mccza.com. Accessed April
29, 2009.
6. Circular 14/95, Data Required as Evidence of Efficacy, Annexure 13, Medicines Con-
trol Council, Department of Health, October 2, 1995.
7. Medicines and Related Substances Control Act, 1965 (Act No. 101 of 1965) as amended
by Act No. 59 of 2002. http://www.mccza.com. Accessed April 29, 2009.
8. Midha KK, Rawson MJ, Hubbard JW. Commentary: The role of metabolites in bioe-
quivalence. Pharm Res 2004; 21:1331–1344.
9. General Information: Medicines Control Council, Department of Health, 2.01 General
Information, February 08, v4, April 2008. http://www.mccza.com/showdocument.
asp?Cat=17&Desc=Guidelines%20-%20Human%20Medicines. Accessed April 30,
2009.
10. Gibaldi M, Perrier D. Pharmacokinetics. 2nd ed. New York: Marcel Dekker, 1982.
11. Amidon GL, Lennernas H, Shah V,et al. A theoretical basis for a biopharmaceu-
tics classification: The correlation of in-vitro drug product dissolution and in-vivo
bioavailability. Pharm Res 1995; 12:413–420.
12. Guidance form Industry, Waiver of the in vivo bioavailability and bioequivalence
studies for immediate-release solid oral dosage forms based on a Biopharmaceu-
tics Classification System, Food and Drug Administration, Rockville, MD, USA, 2000.
http://www.fda.gov/cder/guidance/index.htm. Accessed April 30, 2009.
APPENDIX I
The following were the requirements for proof of efficacy of generic medicines
as stated in
Circular 14/95 issued on 2nd October, 1995
Section 2. Proof of Efficacy May be Submitted by Using One of the Following
Methods, Depending on the Relevancy:
2.1 Bioavailability
2.2 Dissolution
2.3 Disintegration
2.4 Acid neutralizing capacity
2.5 Microbial growth inhibition zones
2.6 Proof of release by membrane diffusion
2.7 Particle size distribution
2.8 Blanching test
2.9 Any other method an applicant wishes to submit, provided the rationale for submit-
ting the particular method is included.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
2.1 Bioavailability
2.1.1 Bioavailability as proof of efficacy may be used in any instance as proof of
efficacy, but must be used in the following cases:
a) for a modified release tablet or capsule dosage form e.g.; slow release or
sustained release.
b) When a monograph in the USP for an active does not include a method
for dissolution.
c) When the active is mentioned in the attached list A, irrespective of a
monograph being available in the USP.
d) Suspensions, except when a monograph for dissolution is available in
the USP for the active in suspension.
e) Exceptions as indicated under alternative methods of proof of efficacy.
2.1.2 Experimental subjects. Generally speaking 12 subjects are required in a
cross-over study for immediate release tablets or capsules, and 20 subjects
for modified release tablets or capsules.
2.1.3 When bioavailability studies presented for registration purposes were
derived from pilot batches, acceptable data derived from production batches
must be submitted before distribution of the production batches can take
place.
Part 2.2.1 of Circular14/95, viz.;
Dissolution as proof of efficacy may be used in the following instances:
a) When a monograph for the active in the USP includes a dissolution require-
ment and the active is not on the attached list A.
b) When a monograph for a combination of actives in the USP includes dissolu-
tion requirement and the actives are not on the attached list A.
c) When a monograph for a combination of actives is not included in the USP,
but monographs for the individual actives with dissolution requirements are
included in the USP, these individual monographs may be used, provided
that the actives are not on the attached list A.
The “attached list A” contained a number of drug substances listed in
alphabetic order and the list was headed as follows:
“MEDICINES CONTAINING THE FOLLOWING CHEMICAL ENTI-
TIES, CERTAIN COMBINATIONS, SPECIFIC GROUPS OF ENTITIES
OR MEDICINES FALLING INTO A SPECIFIC PHARMACOLOGICAL
GROUP, WHICH WILL USUALLY REQUIRE DATA OTHER THAN COM-
PARATIVE DISSOLUTION DATA AS EVIDENCE OF EFFICACY.”
In addition to the above, the following were also stated as being acceptable
data required as evidence of efficacy.
2.3 Disintegration
2.3.1 Disintegration as proof of efficacy may be used in the following instances:
a) Vitamins or vitamins and mineral combinations when a claim is made
as a supplement.
b) Phenolphthalein
c) Sucralfate
2.3.2 The following are guidelines for the submission of disintegration data:
a) The disintegration test included for Nutritional Supplements in the
USP must be used for the vitamins.
b) The general disintegration test included in the USP may be used for the
other substances.
2.4 Acid neutralising capacity
2.4.1 Acid neutralizing capacity may be as proof of efficacy may be used in the
following instances:
For all products with an antacid or acid neutralizing claim.
2.4.2 The following are guidelines for the submission of acid neutralizing data:
The acid neutralizing capacity test included in the USP must be used.
2.5 Microbial growth inhibition zones
2.5.1 Microbial growth inhibition zones as proof of efficacy may be used in the
following instances:
For all products with a bacteriostatic/bactericidal/antiseptic claim.
SPH SPH
IHBK055-08 IHBK055-Kanfer January 6, 2010 20:13 Char Count=
Ricardo Bolaños
Department of Pharmacology, School of Medicine, University of Buenos Aires;
Department of Projects and Plans, Direction of Planification, National
Administration of Drugs, Food and Medical Technology (ANMAT), Ministry of
Health; and Working Group of Bioequivalence, Pan American Network of Drug
Regulatory Harmonization, PAHO, Buenos Aires, Argentina
INTRODUCTION
Current medicine policies, both in developed and underdeveloped countries,
seek the same objectives of efficacy, safety, and quality of accessible products.
However, different historical, social, and economic circumstances in such coun-
tries have resulted in different scenarios associated with their own internal issues
which have been and are being currently resolved according to each particular
prevailing situation.
The map of registration requirements for pharmaceutical products in the
Americas is heterogeneous. Neither registration of innovator products nor that
of noninnovator ones are identical and the documentation requirements for the
registration of either are different among those countries.
There is currently a competitive market between innovator products and
noninnovator products. The noninnovator products involve both generic prod-
ucts and so-called “similar products.” The term “generic product” was initially
used in those countries that recognized the intellectual property of pharmaceuti-
cal products to refer to products registered by competitors once the patent protec-
tion period had expired. Usually generic products are commercialized without
a brand name although sometimes a brand name is used (the brand generics).
The term “similar product” is used in countries where, historically, no patent
protection rights on pharmaceutical products existed. In those cases, different
commercial brands of the same active drug substances are simultaneously com-
mercialized by different pharmaceutical laboratories. Definitions for both generic
and similar products are included in the multisource product definition (1).
Harmonization efforts seek to ensure medicines availability at both
national and international level by agreement of common requirements for regis-
tration, inspection, control, and vigilance. In this context different harmonization
211
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
initiatives has taken place in Europe,a Southeast Asia,b America as well as ICH.c
Particularly, the World Health Organization (WHO)d has provided a number
of guidelines and technical documents related to good manufacturing practices
(GMPs), good clinical practice (GCP), comparator products, essential medicines,
multisource products registration, and so on. WHO has also promoted the
International Conference of Drug Regulatory Authorities (ICDRA). In this con-
text, regional harmonization efforts in the Americas have been carried out by
different economic integration groupse and by PAHO (Pan American Health
Organization) through PANDRH (Pan American Drug Regulatory Harmoniza-
tion). This network grouped Pan-American regulatory authorities since 1997.
Its work has been organized through continuous meetings of different work-
ing groups around the main issues such as GMP, bioavailability/bioequivalence
(BA/BE), GCP, drug registration, phamacopoeia, combat to drug counterfeiting,
GLP, drug classification, botanical products, pharmacovigilance, and vaccines.
Besides the official regulatory authorities, pharmaceutical industry representa-
tives are invited to participate with one observer in each working group. The
Latin-American pharmaceutical companies are thus represented by FIFARMA
(Federación Latinoamericana de la Industria Farmacéutica) and ALIFAR (Asociación
Latinoamericana de Industrias Farmacéuticas). The documents, surveys, and recom-
mendations produced by PANDRH are in the public domain at www.paho.org.
Several papers on drug regulations in the Americas have been recently
published. Homedes and Ugalde (2), reported the results of a preliminary sur-
vey conducted in 10 Latin-American countries. The study aimed to document
the experiences of different countries in defining and implementing generic
drug policies. It focused on determining the cost of registering different types of
pharmaceutical products, the time needed to register them, and uncover incen-
tives that governments have developed to promote the use of multisource drugs
products. The survey instrument was administered in person in Chile, Ecuador,
and Peru and by e-mail in Argentina, Brazil, Bolivia, Colombia, Costa Rica,
a EMEA: European Agency for the Evaluation of Medicinal Products; CADREAC: Collaboration
Agreement of Drug Regulatory European Authorities.
b ASEAN: the Association of Southeast Asian Nations.
c ICH is the International Conference of Harmonization of Technical Requirements for Regis-
tration of Pharmaceuticals for Human Use. It was established in 1990 and is participated by
both regulatory authorities and pharmaceutical industry from the European Union, Japan,
and United States. Today, ICH has a Global Cooperation Group, where different harmoniza-
tion initiatives are represented.
d WHO has an international legal mandate from 191 members to establish global standards for
the promotion and protection of public health.
e The different economic integration groups in the Americas are:
a) TLCN (Tratado de Libre Comercio de Norteamérica): Canada, Estados Unidos, and
Mexico.
b) MERCOSUR: Argentina, Brazil, Paraguay and Uruguay (Chile and Bolivia participate
without being members).
c) SICA (Sistema de la Integración Centroamericana): Guatemala, El Salvador, Honduras,
Nicaragua, and Costa Rica.
d) CAN (Comunidad Andina de Naciones): Bolivia, Colombia, Ecuador, Peru, and Venezuela.
e) CARICOM (Caribbean community): Caribbean Islands.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
REGISTRATION
Background
Since neither physicians nor patients are able to asses the efficacy, safety, and
quality of the medicines that they prescribe and consume, national health author-
ities must establish mechanisms to ensure the various foregoing considerations.
Most countries in the Americas have laws and regulations regarding the registra-
tion, inspection, control, and postmarketing surveillance of medicines. However,
differences can be found, for example in the following:
– Classification.
– Documentation to be submitted for registration.
– Meaning of the terms “new drug substance” and “new drug product.”
– Assessment criteria.
f http://www.paho.org/Spanish/AD/THS/EV/rm-hp.htm.
g http://www.paho.org/Spanish/ad/ths/ev/BE ImpletEstudio04-esp.pdf.
h http://new.paho.org/hq/index.php?option = com content&task = view&id = 1060&Itemid
= 513.
i http://new.paho.org/hq/index.php?option=com content&task=view&id=1052&Itemid=
513&limit=1&limitstart=1.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
j http://www.paho.org/Spanish/AD/THS/EV/rm-hp.htm.
k http://new.paho.org/hq/index.php?option = com content&task = view&id = 1060&Itemid
= 513.
l Author’s note: Drug substance is taken in the context of this article as API (active pharma-
ceutical ingredient).
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
API locally
commercialized?
Yes No
Commercialized Commercialized
outside? outside?
Yes No Yes No
Reference Reference
country? country?
Yes No Yes No
a b c d e f
FIGURE 1 a, b, c: As the drug substance is already being locally commercialized, it will never
be considered a new one. The differences between a, b, and c take into account whether the
drug substance is being commercialized outside the country and the category of those countries
where reference country categories have been locally established.
d, e, f: As the drug substance is not being locally commercialized, some countries will consider
that the drug substance is a new one. However, countries which accept the concept of country
of reference will consider that the drug substance is a new one only in cases e and f.
m Some parts of the original document are reproduced (italics) in the present chapter whereas
others are summarized.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
Product locally
commercialized?
YES NO
Commercialized Commercialized
outside? outside?
NO
YES NO YES
Reference
Reference
country?
country?
YES NO YES NO
A B C D E F
nA nPF nU nP
FIGURE 2 nA, New association (fixed-dose combination); nPF, New pharmaceutical formula-
tion; nU, New use; nP, New potency.
A, B, C: As the drug product is already being locally commercialized, it will never be considered
a new one. The differences between A, B, and C take into account whether the drug product is
being commercialized outside of the country and the category of those countries where reference
countries category has been locally established.
D, E, F: As the drug product is not being locally commercialized, some countries will consider
that the drug product is a new one. However, countries which accept the concept of country of
reference will consider that the drug product is a new one only in cases E and F. Besides, in
cases D, E and F, if the API is an already known one (cases a, b, c, and eventually d of chart
shown in “New Drug Substance” section), documentation requirements for registration of the
new drug product might be different than when the API is unknown.
– New molecules
– New formulations
– New uses
– New dosage form
– New presentation
– New route of administration
– New association
– Generic equivalent
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
– Similar equivalent
– Biologics
– Renewal
The survey implied to answer if the following requirements applied for
registration of the different cases mentioned above:
– General information about the product
– Information about the manufacturing facilities
– Legal documentation supporting the application
– Active Pharmaceutical Ingredient
– Final product
– Biopharmaceutic technical documentation
– Preclinical data
– Clinical data
– Labelling
– Secondary packaging components
– Package Inserts
Observations are summarized as follows mainly taking into account the
information relating to requirements for new molecules (with the knowledge that
new molecules have the strictest requirements of all studied groups):
name (INN or DCI), potency, dosage form, expiration date, storage conditions,
and prescription conditions. Some countries require registration number, instruc-
tions for use, warnings, and contraindications. A fairly low percentage of coun-
tries require manufacturing date, legal representative, name of the responsible
professional, indications, posology, and interactions. Finally, a very small per-
centage of countries require the inclusion of restrictions for use and overdose
data.
Packaging Insert
A package insert for new APIs is required by 69.2% of the countries. A high
percentage of countries require commercial name, generic name, dosage form,
instructions of use, indications, dosage, warnings, contraindications, adverse
reactions, interactions, and overdose in the insert whereas a smaller number of
countries require manufacturer information, expiration date, name of the respon-
sible professional, registration number.
Final Product
A document about the formulation development (44.4% for new molecules),
manufacturing method (61.1% for new molecules), and physicochemical proper-
ties of the excipients (50.0% for new molecules) is required. Formula and storage
conditions are required in 100% of registrations of new molecules. A high per-
centage of countries require stability data and analytical methodology (94.4% for
new molecules) whereas a lower percentage of countries require local analysis
for imported products (50.0% for new molecules).
Preclinical Data
Data relating general and special toxicology, pharmacodynamic, and pharma-
cokinetic studies are required in most countries mainly for new APIs (72.2–
94.4%).
Clinical Data
The percentage of countries requiring clinical data are the following:
◦ New chemical entities: 72.2%
◦ New formulation: 50.0%
◦ New indication: 66.7%
◦ New dosage form: 61.1%
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
n Parts of the original document are reproduced (italics) in the present chapter.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
BIOEQUIVALENCE
Introduction
Bioequivalence requirements for registration of noninnovator products in the
Americas are basically divided into three different approaches as follows, United
States/Canada, Brazil/Mexico, and the rest of the Spanish-speaking countries.
On the one hand, United States and Canada will always require demon-
stration of therapeutic equivalence-employing either in vivo or in vitro
methodology- to allow health authorities to declare interchangeability between
the noninnovator product (the generic product) and the reference product (usu-
ally the innovator product). When a noninnovator product fails to demon-
strate therapeutic equivalence, it will not be declared interchangeable and conse-
quently it will not be registered as a generic product in either the United States
or in Canada.
A different approach is taken in Mexico and Brazil. Both countries have
regulations for the registration of generic products from 1999. Mexico was the
first Latin-American country with the Norma Oficial Mexicana NOM-177-SSA1–
1998 and Brazil was the second with the Ley No 9787 from 10.2.99 and the Res. No
391 from 9.8.99. However, both of them allow sponsors to choose between two
types of registration of noninnovator products, interchangeable generic products
and similar products.
Finally, the rest of the Spanish-speaking countries represent the third
approach. In general, they do not have regulations for the registration of generic
products as such.
However, in some of the countries, a bioequivalence study and/or decla-
ration of interchangeability (through either in vitro or in vivo methodology) is
also required as a condition either for registration or commercialization for some
of the noninnovator products, mainly chosen according to high health risk cri-
teria. For example, the following countries use this type of registration and/or
commercialization requirementso :
r Argentina: Disp. No 3185/1999 from ANMAT.
r Chile: Res. No. 726 and 727/2005 from Health Ministry.
r Colombia: Res. No. 1400/2001 from Health Ministry.
r Costa Rica: Pres. Dec. No.32470-S/2005.
r Cuba: Reg. No. 18-07 from Health Ministry.
r Panama: Res. No. 081/2005 from Health Ministry.
r Venezuela: Normas de la Junta Revisora de Productos Farmacéuticos, from Jan-
uary 1999, Chapter XIV and more recently in Res. No. 38.499/2006 from Health
Ministry.
r Uruguay: Pres. Dec. of Interchangeability, 2008.
In some countries, such as Peru and Ecuador, expert groups are discussing
the way to include therapeutic equivalence studies (either in vitro or in vivo)
requirements into their own regulations.
o The following summary states mainly the first or initial regulations where these requirements
can be found. However, in most cases they have been complemented with additional local
regulations.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
p Usually the term “country of reference” refers to countries with high surveillance health
agencies.
q http://www.paho.org/Spanish/ad/ths/ev/BE ImpletEstudio04-esp.pdf.
r http://new.paho.org/hq/index.php?option=com content&task=view&id=1052&Itemid=
513&limit=1&limitstart=1.
s Some parts of the original document are reproduced (italics) in the present chapter whereas
others are summarized.
t Prioritization criteria are intended to be useful mainly in countries where BE studies are not
included into registration requirements and refer to criteria to choose which APIs have priority
mainly considering sanitary risk.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
Prioritization Criteria
Thirty-five percent of participating countries (∼16% of Pan American countries)
have already established prioritization criteria with variable number of drugs in
positive lists (countries usually list locally prioritized drugs) and this impacts on
66% of Latin-American/Caribbean population.
u There were 20 participant countries of the survey of 46 countries within all the Americas.
Those 20 countries correspond to 93.6% of the total population of the Americas and 18 of
them correspond to 89.8% of the Latin-American and Caribbean population.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
Training
Forty percent of participating countries (∼18% of Pan-American countries)
report having carried out BE training courses and this impacts on 70% of
Latin-American/Caribbean population.
From the date of the survey until the time of writing this chapter, addi-
tional countries have developed new BE regulations, for example, Chile, Costa
Rica, Cuba, Panama, and Uruguay. Hence, since the start of the present survey,
an increasing number of countries have included BE studies as a registration
requirement affecting about 80% of Latin-American population and about 50%
of the population of the Americas.
v Some parts of the original document are reproduced in the present chapter (italics) whereas
others are summarized.
w First version of the document was considered during the IV PANDRH Conference, March
x Taken as an example from API´s listed in: “Multisource (generic) pharmaceutical prod-
ucts: guidelines on registration requirements to establish interchangeability” WHO Technical
Report Series (1996), No. 863, 114–155.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
The same document also shows a list of APIs subject to BE in vivo study
requirements in different countries in the American region.
Product Manufacturer
meets GMP
requirements?
YES NO
Must
Product requires meet
equivalence GMP
study?
YES NO
Go to
Is a hhr Register
API? Office
NO
YES
If DRA requires
BE: Elegible for
Has a valid Ref Prod? Biowaver?
YES NO
YES NO
Has a valid
Proceed as
Ref. Prod ?
hhr API
Apply
BE WHO
study criteria and
YES NO
this
document
y Note of Authors: “Reference product” corresponds to the term “comparator product” as defined
by WHO.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
authorities will accept that the innovator product is the most logical compara-
tor product for a multisource pharmaceutical product because its quality, safety,
and efficacy would have been comprehensively assessed and documented. In
Latin America, however, despite the availability of innovator products in the
local market, national requirements of reliability for a reference product still has
to be assessed. Accordingly, in the light of this complex scenario, the PANDRAH
WG/BE has included a recommended mechanism to help the national health
authority to assess the local particular situation and to establish a reliable refer-
ence product.
z United States, Japan, Sweden, Swizerland, Israel, Canada, Austria, Germany, France, United
Kingdom, The Netherlands, Belgium, Denmark, Spain, and Italy.
aa Australia, Mexico, Brazil, Cuba, Chile, Finland, Hungary, Irland, China, Luxenburg, Norway,
The authors declare that the contents of this chapter represent their
personal point of view without involving any organizations where they are
associated.
ABBREVIATIONS
ACKNOWLEDGMENTS
We acknowledge the dedication and professional work of members of PAN-
DRH Working Groups, Drug Registration and Bioequivalence, as well as their
Coordinators: Esperanza Briceño (Venezuela), Marı́a Teresa Ibarz (Venezuela),
and Justina Molzon (United States). In addition, with special deep grati-
tute and respect, we recognize the inestimable help and support of Rosario
D’Alessio, (PANDRH/PAHO secretariat). We specially acknowledge the con-
tinuous support and expertise from Nelly Marı́n (PANDRH/PAHO secretariat)
and the careful revision of this chapter and precise advice from Horacio
Pappa.
SPH SPH
IHBK055-09 IHBK055-Kanfer January 8, 2010 19:43 Char Count=
REFERENCES
1. Annex 7 “Multisource (generic) pharmaceutical products: guidelines on registration
requirements to establish interchangeability. WHO Technical Report Series, No. 937,
2006.
2. Homedes N, Ugalde A. Multisource drug policies in Latin America: Survey of 10 coun-
tries. Bull World Health Organ 2005; 83(1):64–70.
3. Vacca González CP, Fitzgerald JF, Bermúdez JAZ. Definición de medicamento genérico
¿un fin o un medio? Análisis de la regulación en 14 paı́ses de la Región de las Américas.
Rev Panam Salud Pública/Pan Am J Public Health 2006; 20(5):314–323.
4. Annex 8. Proposal to waive in vivo bioequivalence requirements for WHO Model List
of Essential Medicines immediate-release, solid oral dosage forms. WHO Technical
Report Series, No. 937, 2006.
5. Argentinean regulations relating medicines registration:
r Dec. ANMAT 150/92
r Res. MS No1062/08
r Disp. ANMAT No 4541/08
r Dec. PEN No 253/08
r Res. MS No 102/09
r Disp. ANMAT No 1310/09
Argentinean regulations relating the use of generic names in prescriptions:
r Res. SP No 326/02
r Ley Esp. Med. No 25.649/02
r Dec. No 987/03
Argentinean regulations relating GMP and current further regulations:
r Disp. ANMAT No 2819/04
r Disp. ANMAT No 3477/05
r Disp. ANMAT No 2372/08
SPH SPH
IHBK055-10 IHBK055-Kanfer January 6, 2010 19:39 Char Count=
10 Taiwan
Li-Heng Pao
School of Pharmacy, National Defense Medical Center, Taipei, Taiwan,
Republic of China
Jo-Feng Chi
Bureau of Pharmaceutical Affairs, Department of Health, The Executive Yuan,
Taipei, Taiwan, Republic of China
Oliver Yoa-Pu Hu
National Defense Medical Center, Taipei, Taiwan, Republic of China
INTRODUCTION
The Bureau of Pharmaceutical Affairs (BPA) within the Department of Health
(DOH) is responsible for the regulation of medicinal products in Taiwan. The
mission of the BPA is to ensure that medicinal products that are available for
the people in Taiwan are of the highest quality, safety, and efficacy. On the basis
of BPA statistics, there are over 27,000 active licensed drugs currently available
in the Taiwan market (1). Approximately 20% of Taiwan’s drug products are
imported from abroad, the remainder being manufactured by domestic pharma-
ceutical companies that operate in compliance with current good manufactur-
ing practice (cGMP) standards. Therefore, the need to ensure bioequivalence of
generic drug products with corresponding innovator products is critical in Tai-
wan. The goal of this chapter was to provide an overview of the generic drug
review process and bioequivalence requirements in Taiwan.
Currently the BPA is organized into five sections. Section I is responsible for
the law and regulation of pharmaceutical affairs, pharmacy-related compliance,
and advertising. Section II is involved with the registration of medical devices
and cosmetics. New drug applications (NDAs), clinical trials, and postmarket-
ing surveillance are reviewed and approved according to Section III. Section
IV covers the review of abbreviated new drug applications (ANDAs) and their
approval for marketing whereas Section V deals with registration of biological
and plasma products, radiopharmaceuticals, as well as in vitro diagnostic kits.
The Taiwan Food and Drug administration (TFDA) will be formally inaugurated
Jan. 1, 2010. Additional information on the organization of the BPA can be found
at www.doh.gov.tw.
LEGISLATIVE AND REGULATORY ISSUES
Drug regulation has become more challenging since the advancement of new
technologies, cost pressures, as well as trends of globalization. The drug regu-
latory process and development of the pharmaceutical industry in Taiwan has
changed substantially over the past two decades (2–4). The fundamental law
regulating medicinal products is based on the Law for Control of Medicaments and
Pharmaceutical Firms promulgated in 1970. The Law was extensively revised and
232
SPH SPH
IHBK055-10 IHBK055-Kanfer January 6, 2010 19:39 Char Count=
Taiwan 233
renamed the Law of Pharmaceutical Affairs in 1993. The current Law of Pharmaceu-
tical Affairs, with its subsequent amendments in 2004, is the basic law for the
regulation of medicinal products in Taiwan. Implementation of good manufac-
turing practice (GMP) and cGMP had a dramatic impact on Taiwan’s pharma-
ceutical industry. Before advent and implementation of GMP requirements in
1982, there were over 400 domestic pharmaceutical manufactures. Six years later
in 1988, the number of manufactures was reduced to 230. To further improve and
meet the international standards of domestic production of medicinal products,
cGMP regulations were promulgated in 1999. There are currently approximately
160 cGMP pharmaceutical manufactures in Taiwan. Therefore, when applicants
seek approval of generic products in Taiwan, the drug products must comply
with cGMP standards and requirements.
Following successful implementation of the GMP program in Taiwan,
academia, government, and the pharmaceutical industry recognized that data
on bioavailability and bioequivalence (BA/BE) were critical to further improve
the quality of medicinal products in Taiwan. Consequently, the BPA contracted
university professors in 1984 to draft BA/BE guidelines. Through lengthy dis-
cussions and amendments via discourse with academics, scientists from indus-
try, government officers, as well as overseas Chinese scientists, the first Taiwan
guidelines on BA/BE for generic medicines were issued in 1987. A system of
post-marketing safety (PMS) surveillance for new medicinal products was imple-
mented in 1983. All generic products containing new chemical entities (NCEs)
approved under the PMS surveillance were required to provide BA/BE data
when submitting marketing authorization applications since the implementation
of BA/BE guideline in 1987. Since that time, several revisions of the guidelines
have been made on the basis of current BA/BE guidelines used in various coun-
tries and also on relevant practical situations and issues prevailing in the phar-
maceutical industry in Taiwan.
Currently, “generic substitution” is not compulsory in Taiwan.
The on-site overseas inspection program was established in 2002. Over 100
foreign on-site cGMP inspections in 28 countries have been completed since the
program was implemented in 2003. For imported products, the DOH reviews
the plant master files (PMFs) submitted by the pharmaceutical company as a
substitute for on-site inspection. The requirements for a PMF to be submitted
with marketing authorization applications apply only for those plants without
a PMF approved in Taiwan. The DOH is trying to establish a mutual recogni-
tion policy (PIC/S) amongst countries to ensure the quality and efficacy of for-
eign products and then the submission of the PMF and on-site inspection can
be waived. PIC/GMP standards have been implemented since 2008. All phar-
maceutical manufactures in Taiwan will need to comply with PIC/GMP require-
ments after year 2012. However, translation of the PMF into English requires a
huge effort and is very time consuming. Consideration of the confidentiality of
the PMF submitted to another country is also considered to be an issue of concern
for some pharmaceutical companies in Taiwan.
The PMF for any imported products generally involves the site of manu-
facture and the product dosage form. Marketing authorization applications may
be waived in Taiwan if the imported product consists of the same dosage form
and has been manufactured at the same site with a previously approved PMF.
Taiwan 235
Applicant
ANDA
Application
complete ? No
Yes
Labeling
Validation and specifications Chemistry and Bioequivalence
(PMF/cGMP) Manufacture control
Stability data
and dosage, legal status, permit license number, expiry date, batch number,
name/address of manufacturer or agent, adverse drug reactions, warning, con-
traindications, and storage conditions. The labeling review is based on the ref-
erence drug product labeling to ensure that the essential information already
described is properly labeled.
Chemistry and Manufacture Quality Review Process
Following acceptance for filing by the staff (Section IV), the chemistry and man-
ufacture control and stability of the finished dosage form are reviewed by a BPA
reviewer and the relevant specifications are forwarded to the BFDA, an indepen-
dent institute of the DOH.
Data such as manufacturing process validation, analytical method valida-
tion, and stability of the finished dosage form and PMFs are reviewed by the
BFDA to ensure that the generic product will be manufactured in compliance
with the standards of cGMP and relevant pharmacopoeial specifications when
applicable. In addition, a number of samples of the finished product must also
be supplied for analysis according to its own product specifications by BFDA to
further ensure the quality of the drug.
Bioequivalence Review Process
Subsequently, the BE document is randomly assigned to one external indepen-
dent expert on BA/BE to review. If additional information is required or prob-
lems are raised during the BE review, the reviewer will inform the staff in the
BPA resulting in a document that is issued by the DOH to the applicant who is
required to resolve the deficiency or specified problems and the response time
limit is 70 days. Once the BE review has been completed and all BE requirements
are fulfilled based on the current BE guidelines, an official document, accepting
the BE results, is issued to the applicant by the DOH.
To ensure the quality and credibility of the BE study, the BA/BE inspec-
tion program was established in 2002. During the BE review process, the clinical
and analytical sites are randomly inspected on a case- or problem-oriented basis.
BE studies of imported drug products performed outside Taiwan are currently
acceptable (2008). The certification of foreign BE laboratories and facilities has
not yet been possible. When the review of an ANDA is completed, an official
document is sent to the applicant as notification of marketing approval of the
generic drug product in Taiwan.
BA/BE GUIDELINES IN TAIWAN
Establishing bioequivalence is an important part of the generic review process
and is closely linked to the concepts of essential similarity and product inter-
changeability. Bioequivalence is defined as no significant difference with respect
to the rate and extent of active ingredients or active moiety in pharmaceutical
equivalents or pharmaceutical alternatives that is available at the site of drug
action when given in the same molar dose under similar study conditions. Princi-
pally, in a BE study, pharmacokinetic end points are used to establish equivalence
between the test and reference drug product. If this is not feasible, pharmacody-
namic as well as clinical endpoints may be used only if they can be scientifically
justified.
Additional information can be found at www.doh.gov.tw.
In vivo bioequivalence may be exempted if the drug product meets any of
the following criteria:
SPH SPH
IHBK055-10 IHBK055-Kanfer January 6, 2010 19:39 Char Count=
Taiwan 237
whichever is higher unless otherwise justified. Quality control based on the prod-
uct specification and test method (i.e., potency and content uniformity) and dis-
solution data of the finished products for both reference and test products should
be submitted as well. A side-by-side comparison of the compositions of test and
reference products is recommended. Dissolution testing should be conducted in
accordance with pharmacopoeial requirements. Alternative methods can be con-
sidered with scientific justified.
Study Design
The study protocol should be approved by an ethics review board and written
informed consent should be obtained from all subjects prior to participation in
the study. In general, the typical randomized cross-over design with appropriate
washout periods is recommended for the BE study. Washout periods between
each treatment should be more than five elimination half-lives of the parent drug
or active metabolite. A parallel study design can be used for oral drug products
with a long half-life, which should be justified by the applicant. In general, sub-
jects should fast for at least 10 hours prior to administration of the drug except for
a food-effect BE study. The drug products should be administered with at least
200 mL of water and water is not allowed at least one hour before and after drug
administration. A standard meal should be provided no less than four hours
after drug administration. The subjects should abstain from other medicines or
alcohol for at least two weeks before and during the study.
Single- Versus Multiple-Dose Studies
A single-dose fasting pharmacokinetic study is generally recommended for con-
ventional immediate-release drug products to demonstrate BE. Multiple-dose
studies may be appropriate for the BE of immediate-release dosage form as
described.
Study subjects are patients.
The assay sensitivity precludes the possibility to adequately characterize the
pharmacokinetic of drug after single-dose administration.
Drugs that exhibit nonlinear kinetics following multiple administrations
For modified-release drug products, either multiple-dose or single-dose
studies under both fasting and fed conditions are required. Omission of either
fasting or fed study should be justified by the applicant. If a multiple-dose study
design is employed, attainment of steady state is essential. At least three consec-
utive trough drug concentrations on three consecutive days should be obtained
to demonstrate that steady-state has been reached.
Number of Subjects
A sufficient number of subjects, which is based on sample-size estimation with
appropriate power calculation should be included in the BE study. Generally,
the minimum number of subjects should be not less than 12 for the assessment
of bioequivalence. An add-on study is not encouraged unless this is anticipated
and should be stated clearly in the protocol as well as details of the proposed
statistical treatment.
Study Population
In general, healthy adult (males, females, or both) volunteers should be
employed and the inclusion of subjects from the target population that the drug
SPH SPH
IHBK055-10 IHBK055-Kanfer January 6, 2010 19:39 Char Count=
Taiwan 239
infinity (AUC0–∞ ) are used for the evaluation of BE. For multiple-dose studies,
area under the curve of a dosing interval (AUC ), Cmax , Cmin , average concentra-
tion (Cave ), and fluctuation at steady state should be assessed for BE evaluation.
Parameters such as mean residence time (MRT), kel , and t1/2 are recommended
for submission as reference data. These latter secondary parameters help justify
the consistency and quality of the BE data.
When urine samples are used, the urinary excretion rate and cumulative
amount of parent drug can be used to evaluate BE. All results from the BE
study should be presented clearly and all individual data and results should be
reported including dropouts, subject withdrawals, and exclusion of data. Any
exclusion of data including outliers should be scientifically justified.
Chromatograms of at least one-third of the subjects and chromatograms of
the full validation of the analytical method should be submitted as well.
Statistical Analysis
A 90% confidence interval (CI) or two one-sided test with a significance level
of 5% should be used for assessing BE. Analysis of variance (ANOVA) should
be applied to the study data to determine the 90% CI. In general, logarithmic
transformation should be provided for measures used for BE demonstration. The
pharmacokinetic parameters, except for Tmax , obtained from the study should
be analyzed using ANOVA after logarithmic transformation. The variables such
as subject, period, sequence, and treatment should be included in the model of
ANOVA and the results of the ANOVA analysis should also be reported. In addi-
tion to the 90% CI for BE assessment, a summary of the statistics of all the rele-
vant pharmacokinetic parameters should be given. If a BE dossier is submitted
where the biostudy was performed some years previously, the review is consid-
ered on a case-by-case basis depending on the nature of the drug, drug product,
and scientific evidence.
BE Acceptance Criteria
The acceptable range for the declaration of BE is that the 90% CI of the ratios
of AUC0–∞ and Cmax of the test to reference product should be within 0.8 to
1.25 limits. Under the current BE guidelines, the BE limit remains unchanged for
narrow therapeutic range drugs. For drugs with no safety or efficacy concerns
(i.e., for a drug with a wide therapeutic window or flat dose–toxicity relation-
ship and with clinical justification), a wider range, for example, 0.75 to 1.34, for
Cmax may be acceptable whereas the AUC criterion remains at 0.8 to 1.25. In such
cases, scientific justification should be provided by the applicant and consulta-
tion with the DOH is required. Currently, there is no special provision for the BE
acceptance criteria for highly variable drugs.
Taiwan 241
REFERENCES
1. Bureau of Pharmaceutical Affairs, Department of Health, Executive Yuan, Taiwan,
Republic of China, 2009.
2. Hsieh YY, Hunag WF. The drug regulatory process of the Republic of China. J Clin
Pharmacol 1998; 28:200–203.
3. Hu O YP, Hsiao ML, Liu LL. Drug regulatory process of the Republic of China-Taiwan’s
experiences in bioavailability and bioequivalence. Drug Inf J 1995; 29:1049–1054.
4. Tzou MC, Chi JF, Hu O YP. Generic drug in Taiwan. Regul Aff J 1999; 11:554–560.
5. Guidance for Industry: Bioanalytical method validation, 2001. http://www.fda.gov/
cder/guidance/4252fnl.htm. Accessed May 9, 2009.
6. 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, 1995. http://www.fda.gov/
cder/guidance/cmc5.pdf. Accessed May 9, 2009.
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
11 Turkey
Ilker Kanzik
IDE Pharmaceutical Consultancy Ltd. Co., Istanbul, Turkey
A. Atilla Hincal
IDE Pharmaceutical Consultancy Ltd. Co., Ankara, Turkey
INTRODUCTION
Registration
Marketing authorization (registration decisions) for medicinal products for
human use is issued by the Ministry of Health (MoH), but the entire procedure
is managed by the General Directorate of Drug and Pharmacy (GDDP). On the
other hand, for veterinary medicinal products and dietary supplements such as
vitamins and minerals, herbal extracts, and other substances that are generally
found in foods, the Ministry of Agriculture is the relevant authority.
Turkish licensing regulations (regulation) for all (i.e., innovator/brand and
generic drug products) pharmaceutical products published on January 19, 2005
(1), came into force on June 30, 2005. This new legislation brought Turkish law in
line with that of the European Union (EU) and covered all aspects of the registra-
tion procedure. According to this legislation the MoH will take all appropriate
measures to ensure that the procedure for granting an authorization to place a
medicinal product on the market is completed within 210 days of the submission
of a valid application.
Applicants intending to register any drug product must submit an appli-
cation for authorization to the GDDP. Applications for marketing authorization
of an innovator/brand drug product must be accompanied by the appropriate
fees as well as various documents and particulars, including the manufacturer’s
product development methodology and processes, control methods (sterility
tests, stability tests, etc.), and the results of physicochemical, biological or
microbiological, pharmaco-toxicological tests, and clinical trials (1).
Prescription (generic or innovator/brand) and nonprescription medicines
(over the counter, OTC) follow the same marketing authorization procedure. It
is likely that after initial consideration, additional questions will be put to the
applicant and the answers would then be further considered by the Registration
Approval Committee leading to a final opinion on the application. If an applica-
tion is refused, the reasons for the decision will be set out in full. The applicant
has the right to object in written or orally. A marketing authorization will be
refused if it appears that the medicinal product is harmful under normal condi-
tions of use, that it has no or very little therapeutic effect or that it does not have
the stated composition in terms of quality and quantity. Authorization will also
be refused if the information requested is incomplete or not given. Marketing
242
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
Turkey 243
Abridged Application
For abridged applications, without prejudice to the provisions of the Decree on
the Protection of the Patent Rights dated 24.06.1995 no: 551 (7), applicants shall
not be required to provide the results of toxicological and pharmacological tests
or the results of clinical trials if they can demonstrate: either that the medici-
nal product is similar to a medicinal product authorized in Turkey and that the
holder of the marketing authorization for the original medicinal product has
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
Turkey 245
relating to the trial are conducted, for their compliance with the provisions of
this regulation and other relevant legislations. Depending on the result of the
inspection, the trial may be stopped if necessary by the Ministry.
Definitions
The following definitions are specifically described in the Turkish BE guide-
lines/regulations/laws. However, those BE regulations are relatively old (1994),
hence the relevant definitions in EU Guideline (2001) are also valid for Turkey.
Bioavailability
BA means the rate and extent to which the active substance or its active moiety
is absorbed from a pharmaceutical dosage form and becomes available at the site
of action or biological fluids (usually plasma or plasma) representing the site.
Bioequivalence
Two medicinal products are bioequivalent if they are pharmaceutically equiva-
lent and if their BAs after administration in the same molar dose are similar to
such a degree that their effects, with respect to both efficacy and safety, will be
identical.
Pharmaceutical Equivalence
Medicinal products are pharmaceutically equivalent if they contain the same
amount of the same active substance(s) in the same dosage forms and admin-
istered by the same route and that meet the same or comparable standards.
Therapeutic Equivalence
A medicinal product is therapeutically equivalent with another product if it con-
tains the same active substance or therapeutic moiety and, clinically, shows the
same efficacy and safety as that product, whose efficacy and safety have been
established.
Study Design
The study should be designed in such a way that any formulation effect can
be distinguished from other effects. For oral suspensions and immediate-release
tablets and capsules, a single-dose in vivo fasting study is usually sufficient. To
reduce the variability, a cross-over design is generally the first choice. When
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
Turkey 247
Subjects
Selection of Subjects
The studies should normally be performed using healthy volunteers. The inclu-
sion/exclusion criteria should be clearly stated in the protocol. In general, vol-
unteers suitable for inclusion should be within the age limits (between 18 and
55 years) and of weight within the normal range according to accepted normal
values for the body mass index (BMI), where BMI = mass (kg)/height (m)2 . Sub-
jects should be asked for informed consent and thereafter screened for suitability
by means of clinical laboratory tests, an extensive review of medical history, and
a comprehensive medical examination with respect to inclusion and exclusion
criteria. Subjects should preferably be nonsmokers and without a history of alco-
hol or drug abuse. If moderate smokers are included (less than 10 cigarettes per
day) they should be identified as such and the consequences for the study results
should be discussed.
Genetic Phenotyping
Phenotyping and/or genotyping of subjects should be considered for
exploratory BA studies and all studies by using parallel group design. It may also
be considered in cross-over studies (e.g., BE, dose proportionality, food interac-
tion studies, etc.) for safety or pharmacokinetic reasons. If a drug is known to be
subject to major genetic polymorphism, studies could be performed in panels of
subjects of known phenotype or genotype for the polymorphism in question.
Characteristics To Be Investigated
Moieties to be measured in BE studies are active drug substances or the active
moiety in the administered dosage form (parent drug). In some situations, how-
ever, measurements of an active or inactive metabolite may be necessary instead
of the parent compound. Such situations include cases where the use of a metabo-
lite may be advantageous to determine the extent of drug input, for example, if
the concentration of the active substance is too low to be accurately measured in
the biological matrix (e.g., major difficulty in analytical method, product unsta-
ble in the biological matrix or half-life of the parent compound too short) thus
giving rise to significant variability.
In BA studies, the shape of, and the area under the plasma concentration
versus time curves are mostly used to assess extent and rate of absorption. The
use of urine excretion data may be advantageous in determining the extent of
drug input in case of products predominately excreted renally, but has to be jus-
tified when used to estimate the rate of absorption. Sampling points or periods
should be chosen such that the time–drug concentration profile is adequately
defined so as to allow the estimation of relevant parameters.
From the primary results, the desirable BA characteristics are estimated,
namely AUCt , AUC∞ , Cmax , tmax , Aet , Ae∞ as appropriate, or any other justi-
fiable characteristics. The method of estimating AUC values should be speci-
fied. For additional information t1/2 and MRT can be estimated. For studies at
steady state, AUC , Cmax , Cmin , and fluctuation [(Cmax − Cmin )/Cav ] should be
provided.
Pharmacodynamic studies are not recommended for orally administered
drugs when the drug is absorbed into the systemic circulation and a pharmacoki-
netic approach can be used to assess systemic exposure and establish BE. How-
ever, in some cases the quantitative measurement of a drug and/or metabolite
in plasma or urine cannot be made with sufficient accuracy and/or reproducibil-
ity. In those cases, studies in healthy volunteers or patients using pharmacody-
namic parameters may be used for establishing equivalence between the test and
reference products. Demonstration of a dose–response relationship may become
necessary. Measurements should be made with sufficient frequency to permit
a reasonable estimate of the total area under the effect–time curve. The baseline
values in each period should be comparable and the complete effect curve should
remain below the maximum physiological response. The methodology used for
carrying out the study should be validated for precision, accuracy, specificity
and reproducibility. Nonresponders should be excluded from the study by prior
screening. A correction for potential nonlinearity of the relationship between the
dose and area under the effect–time curve should be made and also baseline cor-
rections should be considered during data analysis.
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
Turkey 249
Batch Size
In the case of oral solid forms for systemic action, the test product should usu-
ally originate from a batch of at least 1/10 of production scale or 100,000 units,
whichever is greater, unless otherwise justified. The production of batches used
should provide a high level of assurance that the product and process will be
feasible on an industrial scale; in case of a production batch smaller than 100,000
units, a full production batch will be required. If the product is subjected to fur-
ther scale-up this should be properly validated.
Samples of the product from full production batches should be compared
with those of the test batch, and should show similar in vitro dissolution profiles
when employing suitable dissolution test conditions.[see Appendix II of Note for
Guidance on the Investigation of Bioavailability and Bioequivalence, 2001 (9)].
The study sponsor must retain a sufficient number of all investigational
product samples used in the study for one year after the product’s shelf life or
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
two years after completion of the trial or until approval whichever is longer to
allow re-testing, if it is requested by the authorities.
Data Analysis
The main goals and objectives of a BE study is to demonstrate equivalency within
a clinically significant acceptance range (80–125%) and to limit the risk of false
acceptance decisions of BE. In certain cases, however, a wider interval may be
acceptable. The interval must be prospectively defined, for example, 0.75 to 1.33,
and justified addressing, in particular any safety or efficacy concerns for patients
switched between formulations. The possibility offered here by the guideline to
widen the acceptance range of 0.80 to 1.25 for the ratio of Cmax (not for AUC)
should be considered exceptional and limited to a small widening (0.75−1.33).
Furthermore, this possibility is restricted to those products for which at least one
of the following criteria applies:
1. Data regarding pharmacokinetic/pharmacodynamic (PK/PD) relationships
for safety and efficacy are adequate to demonstrate that the proposed wider
acceptance range for Cmax does not affect pharmacodynamics in a clinically
significant way.
2. If pharmacokinetic/pharmacodynamic data are either inconclusive or not
available, clinical safety and efficacy data may still be used for the same pur-
pose, but these data should be specific for the compound to be studied and
persuasive.
3. The reference product has a highly variable within-subject BA.
A post hoc justification of an acceptance range wider than defined in the
protocol is not acceptable. Information that would be required to justify results
lying outside the conventional acceptance range at the post hoc stage should
be utilized at the planning stage, either for a scientific justification of a wider
acceptance range for Cmax , or for selecting an experimental approach that allows
the assessment of different sources of variability.
When a parametric approach is used, the statistical method for testing rela-
tive BA (e.g., BE) should be based upon the 90% confidence interval for the ratio
of the population means (test/reference), for the parameters under considera-
tion. This method is equivalent to the corresponding two one-sided test proce-
dures with the null hypothesis of bioinequivalence at the 5% significance level.
In the case of concentration or concentration-related characteristics (e.g., AUC)
the data should be transformed prior to analysis using a logarithmic transforma-
tion. In the parametric approach, if it is doubtful to assume log normal (AUC,
Cmax ) or normal distribution (tmax ), a nonparametric approach is recommended.
This approach may also be selected as a general statistical approach to evaluate
all BA characteristics during a specific investigation.
Currently, Turkey follows this approach, even though it is not mentioned
in its regulation.
In Vitro Dissolution
In vitro dissolution test data obtained with the batches of test and reference
preparations that were used in the BA and BE studies should always be reported.
The similarity of dissolution profiles between the test product and reference
product should be demonstrated in each of the three buffers within the range
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
Turkey 251
Reporting of Results
The report of a BA or a BE study should contain complete documentation includ-
ing the study protocol, conduct and evaluation and confirmation (QA) that the
study was in compliance with GCP rules. The authenticity of the whole of the
report is attested by the signature of the principal investigator. Furthermore,
responsible person(s)/investigator involved with any particular section of the
study should sign their respective sections of the report. Names and affiliations
of the responsible person(s)/investigator, site of the study and duration should
be stated. The names and batch numbers of the products used in the study as
well as the composition(s), finished product specifications and comparative dis-
solution profiles should be provided. In addition, the applicant should submit a
signed statement confirming that the test product is the same as the one that is
submitted for registration.
All results should be clearly presented and the method used to derive the
pharmacokinetic parameters (e.g., AUC) from the raw data should be specified.
If pharmacokinetic models are used to evaluate the parameters, the model and
computing procedure used should be justified. The reason for not submitting any
data should be explained. All individual subject data should be included and
individual plasma concentration/time curves presented on a linear/linear and
log/linear scale. All the data from subjects who dropped-out should be included.
Dropouts and withdrawal of subjects should be fully documented and accounted
for. A representative number of chromatograms or other raw data should be
included covering the whole concentration range for all, standard and quality
control samples as well as the specimens analyzed together with the analytical
validation report. Currently, the MoH requests 100% of the chromatograms.
Steady-State Studies
There are no specific recommendations for this type of study. However, the EU
guidance on Modified Release Oral and Transdermal Dosage Forms: Section II:
SPH SPH
IHBK055-11 IHBK055-Kanfer December 28, 2009 12:21 Char Count=
(Pharmacokinetic and Clinical Evaluation), 1999 (10), is the guideline that is fol-
lowed by the Turkish MoH.
Outliers
Only data for clinically proven outliers can be excluded from the statistical eval-
uations (11,12).
Add-On Studies
Add-on studies are not permitted.
Turkey 253
Suprabioavailability
If Suprabioavailability is found, that is, if the new product displays an extent of
absorption appreciably larger than the approved product, reformulation to lower
dosage strength should be considered. In this case, the biopharmaceutical devel-
opment should be reported and a final comparative BA study of the reformulated
new product with the old approved product should be submitted.
In case reformulation is not carried out the dosage recommendations for
the suprabioavailable product will have to be supported by clinical studies. Such
a pharmaceutical product should not be accepted as therapeutically equivalent
to the existing reference product. If marketing authorization is obtained, the new
product may be considered as a new medicinal product.
Finally, it is emphasized that where no specific recommendations are pro-
vided for in the Turkish guidelines, the Turkish MoH requests sponsors to follow
current EU guidelines and recommendations (see Chapter 5 of this book), viz.:
Note for Guidance on the Investigation of Bioavailability and Bioequivalence,
2001; “Questions & Answers on the Bioavailability and Bioequivalence Guide-
line, 2006 and Note for Guidance on Modified Release and Transdermal Dosage
forms: Section II (Pharmacokinetic and Clinical Evaluation), 2000.
REFERENCES
1. Regulation on Licensing for Medicinal Products for Human Use, Official Gazetta No:
25725/19 January 2005.
2. The European Parliament and the Council of the European Union. Directive
2001/83/EC of the European Parliament and of the Council of November 6, 2001
on the Community code relating to medicinal products for human use, CPMP/EWP/
QWP/1401/98. Accessed July 26, 2001.
3. Decision No 1/95 of the EC-Turkey Association Council of 22 December 1995 on
implementing the final phase of the Customs Union (96/142/EC). http://www.
mfa.gov.tr/data/AB/EUAssociationCouncilDecision195CustomsUnionDecision.pdf.
Accessed December 22, 1995.
4. Decision No 2/97 of the EC-Turkey Customs Cooperation Committee. http://www.
avrupa.info.tr/Files/File/RECOURCE CENTRE/key links/DECISION No19.doc.
Accessed May 30, 1997.
5. GATT-TRIPS Implementation. http://www.wto.org. Accessed April 1994.
6. Regulation on the Evaluation of Bioavailability and Bioequivalence of Medicinal
Products Official Gazetta of Turkey No: 21942. Accessed May 27, 1994.
7. Decree on the Protection of the Patent Rights dated 24.06.1995 no: 551; http://
www.tpe.gov.tr/portal/default.jsp. Accessed December 7, 1995.
8. Regulations Regarding Clinical Trials. Official Gaetta of Turkey No: 27089,
December 23, 2008
9. EU Note for Guidance on the Investigation of Bioavailability and Bioequivalence.
CPMP/EWP/1401/98, 2001.
10. EU Note for Guidance on Modified Release Oral and Transdermal Dosage Forms:
Section II: (Pharmacokinetic and Clinical Evaluation). CPMP/EWP/280/96, 1999.
11. Turkish MoH Communication no. 058123 dated 16 November 2006.
12. Turkish MoH Communication no. 038031, June 10, 2008.
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
INTRODUCTION
All prescription and over-the-counter generic drugs marketed in the United
States must meet standards established by the U.S. Food and Drug Administra-
tion (FDA). In approving a new generic drug for marketing, the FDA concludes
that the generic product is therapeutically equivalent to its corresponding ref-
erence product (usually the innovator product, but sometimes another generic
product if the innovator product was withdrawn). The FDA believes that thera-
peutically equivalent drug products can be substituted with the full expectation
that both products will produce the same clinical response (1). A generic drug is
approved by the FDA if it is (i) pharmaceutically equivalent to an approved safe
and effective reference product in that it (a) contains identical amounts of the
same active drug ingredient in the same dosage form and route of administra-
tion, and (b) meets compendial or other applicable standards of strength, quality,
purity, and identify; (ii) bioequivalent to the reference product in that it (a) does
not present a known or potential bioequivalence problem, and it meets an accept-
able in vitro standard (usually dissolution testing), or (b) if it does present such a
known or potential problem, it is shown to meet an appropriate bioequivalence
standard; (iii) adequately labeled; and (iv) manufactured in compliance with cur-
rent good manufacturing practice regulations (1). It is important to note that the
regulatory oversight of generic drug chemistry, manufacturing, and controls is
identical to that imposed upon innovator drug products (2).
BIOEQUIVALENCE
No topic seems so simple but stimulates such intense controversy and misunder-
standing as the topic of bioequivalence. The apparent simplicity of comparing
in vivo performance of two drug products is an illusion that is quickly dispelled
when one considers the difficulties and general public misunderstanding of
the accepted regulatory methodology. In the United States, one often hears
members of the public and medical experts alike stating various opinions on
the unacceptability of approved generic drug products based on misconceptions
regarding the determination of therapeutic equivalence of these products to
the approved reference. These misconceptions include the belief that the U.S.
FDA approves generic products that have mean differences from the reference
product of 20% to 25% and that generic products can differ from each other by
as much as 45%. In addition, some incorrectly assume that, since most bioequiv-
alence testing is carried out in normal volunteers, it does not adequately reflect
bioequivalence and therefore therapeutic equivalence in patients. When the
254
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
regulations found in Title 21 of the Code of Federal Regulations Part 300, Sub-
chapter D: Drugs for Human Use (21 CFR Part 300).
Introduction
Statistical evaluation of bioequivalence studies of systemically active drugs is
based on analysis of drug blood or plasma/serum concentration data. The area
under the plasma concentration versus time curve (AUC) is used as an index
of the extent of drug absorption. Generally, both AUC determined until the
last measurable blood sampling time (AUC0–t ) and AUC extrapolated to infin-
ity (AUC∞ ) are evaluated. Drug peak plasma concentration (Cmax ) is used as an
index of the rate of drug absorption.
(or 75/75–125) rule was added to the criteria. According to the 75/75 rule, the
test/reference ratios of AUC and Cmax had to be within 0.75 to 1.25 for at least
75% of the subjects. This was an attempt to consider individual variability in rate
and extent of absorption. In the early 1980s, the power approach was applied
to AUC and Cmax parameters in conjunction with the 75/75 rule. The power
approach consisted of two statistical tests: (i) a test of the null hypothesis of no
difference between formulations using the F test; and (ii) the evaluation of the
power of a test to detect a 20% mean difference in treatments.
Statistically, the power approach and the 75/75 rule have poor perfor-
mance, and the FDA discontinued the use of these methods in 1986. The prob-
lems with both the 75/75 rule and power approach methods arose from the fact
that they were based on the conventional null hypothesis test of no difference.
Conventional hypothesis testing does not assess the evidence in favor of the con-
clusion that the test and reference means are equivalent, but rather assesses the
evidence in favor of a conclusion that the test and reference means are different,
which is not the question of interest in bioequivalence analysis (20- -22). That is,
the objective of bioequivalence analysis is to establish whether the test and ref-
erence means are equivalent—in other words, is the difference between the two
means an acceptable difference?
proportionally with an increase in dose (39). For such drugs, small differences
in the rate or extent of absorption can potentially have substantial effects on the
AUC (40). Thus, using the highest strength in bioequivalence studies, or, in some
cases, the highest starting dose—so that drug pharmacokinetics are potentially in
the “nonlinear range” ensures that a generic formulation will not pass bioequiv-
alence acceptance criteria unless it is formulated to provide nearly the same rate
and extent of exposure as the corresponding reference product. For drugs for
which rate and/or extent of absorption increases less than proportionally with
an increase in dose (41), the bioequivalence study will be most discriminating
if conducted at the lowest strength or, if only one strength is marketed, at the
lowest recommended dose.
product must be given on an empty stomach for reasons of safety (45) or efficacy
(46), then the FDA may conclude that bioequivalence need only be determined
under fasting conditions.
In very few cases, bioequivalence is evaluated only under fed conditions
because there are safety concerns associated with administration of the product
on an empty stomach (47).
Pharmacokinetic Clinical/PD
Dosage Form Measurement Measurement
Performance
Dose ln Dose
measures. Figure 2 shows that the blood concentration of a drug directly reflects
the amount of drug delivered from the dosage form.
Most bioequivalence studies submitted to the FDA are based on mea-
suring drug concentrations in plasma. In certain cases, whole blood or serum
may be more appropriate for analysis. Measurement of only the parent drug
released from the dosage form, rather than a metabolite, is generally recom-
mended because the concentration–time profile of the parent drug is more sen-
sitive to formulation performance than a metabolite, which is more reflective
of metabolite formation, distribution, and elimination (30). Measurement of a
metabolite may be preferred when parent drug concentrations are too low to
permit reliable measurement. In this case, the metabolite data are subjected to a
confidence interval approach for bioequivalence demonstration. Both the parent
and metabolite are measured in cases where the metabolite is formed by presys-
temic or first-pass metabolism and contributes meaningfully to safety and effi-
cacy. In this case, only the parent drug data are analyzed using the confidence
interval approach. The metabolite data are not subjected to confidence interval
analysis but rather used to provide supportive evidence of comparable therapeu-
tic outcome.
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
Plasma Concentration
R1
R2
D1 D2
Dose
FIGURE 2 The blood concentration of a drug directly reflects the amount of drug delivered from
the dosage form. The corresponding responses over a wide range of doses will be of adequate
sensitivity to detect differences in bioavailability between two formulations. This is illustrated for
two widely different doses, D 1 and D 2 . Any differences in dosage form performance are reflected
directly by changes in blood concentration (R 1 and R 2 ).
R2
Clinical/PD Response
R1
D1 D2
Log Dose
small changes in dose. A dose that is too high will produce a minimal response
at the plateau phase of the dose–response curve, such that even large differences
in dose will show little or no change in pharmacodynamic effect. A pharma-
codynamic study can be conducted in healthy subjects. The pharmacodynamic
response selected should directly reflect dosage form performance but may not
necessarily directly reflect therapeutic efficacy.
The FDA accepts bioequivalence studies with pharmacodynamic end-
points for locally-acting drug products. To be adequately sensitive to distinguish
between two products that are not bioequivalent, the dose used in the pivotal
bioequivalence study should be on the linear portion of the dose–response curve.
In such cases, it is necessary to conduct a pilot pharmacodynamic study by using
the reference product to determine the optimal dose for the pivotal bioequiv-
alence study. Topical corticosteroids are examples of a drug product class for
which the pharmacodynamic approach is suitable (52). In this case, the pharma-
codynamic endpoint is based on the ability of corticosteroids to produce blanch-
ing or vasoconstriction in the microvasculature of the skin. Acarbose is another
example of a drug product for which bioequivalence can be determined using
a pharmacodynamic approach (53). Acarbose lowers blood glucose by inhibit-
ing the activity of ␣-glucosidase within the GI tract following ingestion of food
or other sources of sugar. In this case, the pharmacodynamic endpoint is based
upon the ability of acarbose to lower serum glucose after administration of a
sucrose load
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
Introduction
Under certain circumstances, product quality bioavailability and bioequivalence
can be documented using in vitro approaches (58). In vitro dissolution testing
to document bioequivalence for nonbioproblem DESI drugs remains acceptable.
In vitro dissolution characterization is encouraged for all product formulations
investigated, including prototype formulations, particularly if in vivo absorp-
tion characteristics are well-defined for the different product formulations. Such
efforts may enable the establishment of an in vitro–in vivo correlation. When
an in vitro–in vivo correlation is available (18), the in vitro test can serve as an
indicator of how the product will perform in vivo.
(T − R )2
H0 : > (1)
WR
2
(T − R )2
H1 : ≤ (2)
WR
2
where T and R are the averages of the log-transformed measures Cmax and
AUC for the test and reference products, respectively; usually testing is done at
level ␣ = 0.05; and is the scaled average bioequivalence limit. Furthermore,
(ln )2
= (3)
W0
2
where is 1.25, the usual average BE upper limit for the untransformed
test/reference ratio of geometric means, and W0 = 0.25. Rejection of the null
hypothesis H0 supports the conclusion of equivalence.
2 2
A 95% upper confidence bound for YT − YR /sWR ≤ determined in
a BE study must be ≤, or equivalently, a 95% upper confidence bound for
2
YT − YR − sWR2
must be ≤0. The scaling is mixed. If sWR is less than 0.294,
then the two one-sided tests procedure is used to determine bioequivalence. If
sWR is greater than or equal to 0.294, then the reference-scaled procedure is used
to determine bioequivalence. The value of 0.294 is set by the FDA. Additionally,
the point estimate (test/reference geometric mean ratio) must fall within [0.80,
1.25].
Thus there are two parts to the proposed bioequivalence criteria for highly
variable drugs, the scaled average bioequivalence evaluation and a point esti-
mate constraint. The test product must pass both conditions before it is judged
bioequivalent to the reference product.
The FDA believes that the reference-scaled average bioequivalence
approach addresses many of the concerns about the bioequivalence of highly
variable drugs that have been raised for the past several years. The approach
adjusts the bioequivalence limits of highly variable drugs/products by scaling to
the within-subject variability of the reference product in the study. For drugs and
products that are highly variable, reference-scaling effectively decreases the sam-
ple size needed for demonstrating bioequivalence. The additional requirement of
a point-estimate constraint will impose a limit on the difference between the test
and reference means, thereby eliminating the potential that a test product would
enter the market based on a bioequivalence study with a large mean difference.
F1
F2
F3
F4
F5
F6
F7
0.80 1.25
T/R
FIGURE 4 Hypothetical bioequivalence study results for formulations F1 through F7 illustrate
various scenarios of passing and failing bioequivalence criteria. The width of each 90% confi-
dence interval (CI) is shown as a bar, although in actuality, the log-transformed test/reference
(T/R) ratios are distributed as a bell-shaped curve. F1 and F2 represent results of studies in
which the 90% CIs of the test/reference ratios (T/R) fall between 0.80 and 1.25 (pass bioequiv-
alence criteria). For F1, the ratio of T/R means (point estimate) is near 1.00. For F2, the point
estimate is less than 1.00, but because of low variability, the 90% CI of T/R ratios still falls within
acceptable limits. F3 through F7 show ways in which studies fail to pass CI criteria. With F3, the
point estimate is near 1.00, but because of high variability, the 90% CI is very wide and the drug
does not pass bioequivalence criteria. F3 may pass CI criteria if the number of study subjects is
increased. By contrast, F4 through F7 have variability comparable to F1. F4 represents a failure
on the low side (T is less bioavailable than R), and F5 represents a failure on the high side (R is
less bioavailable than T). Since the point estimates for F3 and F4 are still within the 0.8 to 1.25
range, these formulations may also meet CI criteria if a greater number of subjects are dosed.
F6 does not meet the upper bound of the 90% CI, and the point estimate exceeds 1.25. For F7,
the entire CI is outside the acceptance criteria (bioinequivalence). Formulations F6 and F7 are
so different from the reference that both will still fail CI criteria even if the number of subjects is
increased.
(labeled F1 through F7). For simplicity, the width of the 90% confidence inter-
val is shown as a bar, although it is important to remember that the results are
truly not an even distribution but a normal or log-normal distribution. The log-
transformed test/reference ratios from a bioequivalence study are distributed as
a bell-shaped curve, with most of the subjects’ ratios centered around the cen-
ter or mean, and fewer subjects’ ratios falling at the edges. The top bar in Fig-
ure 4 (F1) represents a study with a 90% confidence interval of the test to refer-
ence ratio falling between the limits of 0.80 to 1.25 and the test/reference ratios
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
centered around 1.00. This is what most applicants would like to achieve with
the to-be-marketed formulation for a given product. The second bar (F2) also
represents a 90% confidence interval of test/reference ratios falling within 0.8 to
1.25. Although the mean test/reference ratio is less than 1.00, the variability is
very low with the result that this product also meets the 90% confidence inter-
val criteria. The remaining bars in Figure 4 show various scenarios of failure to
demonstrate bioequivalence. The third bar (F3) from the top depicts a situation
where the test/reference ratios are still centered around 1.00, but because of high
variability and probably inadequate sample size, the 90% confidence interval is
very wide. This example illustrates how highly variable drugs often need more
subjects to attain sufficient statistical power to pass bioequivalence criteria. It is
very likely that a new study on the same formulation would pass if more sub-
jects were enrolled, thereby increasing the power of the study and decreasing the
resulting width of the 90% confidence interval. The next two bars (F4 and F5)
show results of studies which fail to meet bioequivalence criteria, one a failure
on the low side (test product has lower bioavailability than reference product),
the other a failure on the high side (reference product has lower bioavailability
than test product). These two formulations have comparable variability to the
formulation that passed (F1), but fail because there is a difference between the
test and reference formulations. Because the ratio of means, or point estimate, is
still within the 0.80 to 1.25 limits in each of these two cases, it is also possible
that these two formulations may pass another study if many more subjects were
enrolled. The product represented by the bar that is second from the bottom (F6)
does not meet the upper bound of the 90% confidence interval, and also the point
estimate exceeds 1.25. It is likely that this product will not pass even if the power
of the study is increased by enrolling more subjects. The bottom bar (F7) repre-
sents a very extreme case in which the entire confidence interval is outside the
acceptance criteria. In this extreme case, the two products are bioinequivalent.
The two products are so different that it is highly improbable that repeat studies
would ever demonstrate bioequivalence.
In bioequivalence studies of generic products, the most common reason for
failure is that the study was underpowered with respect to the number of subjects
in the dataset. The width of the confidence interval is controlled by the number
of subjects and by the variability of the pharmacokinetic measures. Studies may
be underpowered for various reasons. The applicant may have failed to enroll an
adequate number of subjects. There may be an excessive number of withdrawals,
or there may be missing data because of lost samples. Sometimes a study may fail
because of subjects who appear to have an aberrant response on a given dosing
day (66). For example, noncompliant subjects may cause the study to fail. The
FDA discourages deletion of outlier values, particularly for nonreplicated study
designs (25).
a drug is safe for use and whether such drug is effective must be submitted.
Similar language was not included in Section 505(j), the section covering the sub-
mission of ANDAs. Therefore, generic firms for many years interpreted this lan-
guage to mean that failed bioequivalence studies did not have to be submitted in
their ANDAs. In November of 2000, the Advisory Committee for Pharmaceutical
Science recommended that generic applicants submit to the FDA results of all
bioequivalence studies on the to-be-marketed (“final”) formulations (67). The
committee expressed the opinion that applicants should submit the results of
failed bioequivalence studies as complete summaries, further suggesting that the
FDA should do a brief, but careful examination to identify potential problems
worthy of requesting additional information.
SUMMARY
Current bioequivalence methods in the United States and other countries are
designed to provide assurance of therapeutic equivalence of all generic drug
products with their innovator counterparts. The sole objective of bioequiva-
lence testing is to measure and compare formulation performance between two
or more pharmaceutically equivalent drug products. For generic drugs to be
approved in the United States, they must be pharmaceutically equivalent and
bioequivalent to be considered therapeutically equivalent and therefore approv-
able. In the United States, a mechanism for submitting ANDAs for generic prod-
ucts was initiated in 1962 and expanded by the Hatch–Waxman amendment of
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
that rate and extent of drug exposure from generic drugs differ very little from
that of their corresponding innovator counterparts (70–72). The FDA believes
that a health care provider can substitute an approved generic product for the
brand product with assurance that the two products will produce an equivalent
therapeutic effect in each patient.
REFERENCES
1. Approved Products with Therapeutic Equivalence Evaluations, 29th ed. Washing-
ton, DC: US Department of Health and Human Services, Public Health Service,
Food and Drug Administration, Center for Drug Evaluation and Research, Office of
Pharmaceutical Sciences, Office of Generic Drugs, 2009. http://www.fda.gov/cder/
orange/obannual.pdf. Accessed July 3, 2009.
2. Current Good Manufacturing Practice for Finished Pharmaceuticals. Title 21 Code
of Federal Regulations. Part 211. Washington, DC: U.S. Government Printing Office.
April 1, 2008.
3. Kefauver-Harris Amendments, Public Law (PL) 87–781, 87th Congress, October. 10,
1962.
4. Drug Efficacy Study: A Report to the Commissioner of Food and Drugs, National
Academy of Sciences. Washington, DC: National Research Council, 1969.
5. 34 Fed Regist 2673, February 27, 1969.
6. 35 Fed Regist 6574, April 24, 1970.
7. 35 Fed Regist 11273, July 14, 1970.
8. Glazko AJ, Kinkel AW, Alegnani WC, et al. An evaluation of the absorption char-
acteristics of different chloramphenicol preparations in normal human subjects. Clin
Pharmacol Ther 1968; 9:472–483.
9. Barr WH, Gerbracht LM, Letcher K, et al. Assessment of the biologic availability of
tetracycline products in man. Clin Pharmacol Ther 1972; 13:97–108.
10. Chiou WL. Determination of physiological availability of commercial phenylbuta-
zone preparations. Clin Pharmacol Ther 1972; 12:296–299.
11. Blair DC, Barnes RW, Wildner EL, et al. Biologic availability of oxytetracycline HCl
capsules: A comparison of all manufacturing sources supplying the United States
market. JAMA 1971; 215:251–254.
12. Lindenbaum J, Mellow MH, Blackstone MO, et al. Variations in biological availability
of digoxin from four preparations. N Eng J Med 1971; 285:1344–1347.
13. Office of Technology Assessment. Drug Bioequivalence. A Report of the Office of
Technology Assessment Drug Bioequivalence Study Panel. Washington, DC: U.S.
Government Printing Office, 1974.
14. Office of Technology Assessment. Scientific Commentary: Drug bioequivalence. J
Pharmacokinet Biopharm 1974; 2:433–466.
15. 42 Fed Regist 1648, January 7, 1977.
16. 57 Fed Regist 17950, April 28, 1992.
17. Drug Price Competition and Patent Term Restoration Act of 1984, Public Law 98–417,
98 Stat 1585–1605, September 24, 1984.
18. 57 Fed Regist 17998, April 28, 1992.
19. Dighe SV, Adams WP. Bioequivalence: A United States regulatory perspective. In:
Welling PG, Tse FLS, Dighe SV, eds. Pharmaceutical Bioequivalence. New York, NY:
Marcel Dekker, Inc., 1991:347–380.
20. Schuirmann DJ. A comparison of the two one-sided tests procedure and the power
approach for assessing the equivalence of average bioavailability. J Pharmacokinet
Biopharm 1987; 15:657–680.
21. Westlake WI. Bioequivalence testing: A need to rethink. Biometrics 1981; 37:589–594.
22. Schuirmann D. Statistical evaluation of bioequivalence studies. In: Sharma KN,
Sharma KK, Sen P, eds. Generic Drugs, Bioequivalence and Pharmacokinetics. New
Delhi, India: University College of Medical Sciences, 1990: 119–125.
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
23. Proceedings of the September 26, 1991, meeting of the Generic Drugs Advisory Com-
mittee. Available from Division of Dockets Management, Food and Drug Administra-
tion, 5630 Fishers Lane, Room 1061 (HFA-305), Rockville, MD, 20852.
24. Bolton S. Statistical considerations for establishing bioequivalence. In: Shargel L, Kan-
fer I, eds. Generic Drug Product Development – Solid Oral Dosage Forms. New York,
NY: Marcel Dekker, Inc., 2005:257–279.
25. US Dept of Health and Human Services, Food and Drug Administration, Center
for Drug Evaluation and Research. Guidance for Industry: Statistical Approaches
to Establishing Bioequivalence, January 2001. http://www.fda.gov/downloads/
Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070244.pdf.
Accessed July 3, 2009.
26. Endrenyi L, Fritsch S, Yan W. Cmax/AUC is a clearer measure than Cmax for absorp-
tion rates in investigations of bioequivalence. Int J Clin Pharmacol Ther Toxicol 1991;
29:394–399.
27. Duquesnoy C, Lacey LF, Keene ON, et al. Evaluation of partial AUCs as indirect mea-
sures of rate of drug absorption in comparative pharmacokinetic studies. Eur J Pharm
Sci 1998; 6:259–263.
28. Tozer TN, Bois FY, Hauck WW, et al. Absorption rate vs. exposure: which is more
useful for bioequivalence testing? Pharm Res 1996; 13:453–456.
29. Basson RP, Ghosh A, Cerimele BJ, et al. Why rate of absorption inferences in
single dose bioequivalence studies are often inappropriate. Pharm Res 1998; 15:
276–279.
30. US Dept of Health and Human Services, Food and Drug Administration,
Center for Drug Evaluation and Research. Guidance for Industry: Bioavail-
ability and Bioequivalence Studies for Orally Administered Drug Products –
General Considerations, March 2003. http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/ucm070124.pdf. Accessed
July 3, 2009.
31. FDA Response to Docket No. 0lP-0546/PSAl& SUP1, November 21, 2003.
http://www.fda.gov/ohrms/dockets/dailys/03/dec03/121003/01p-0546-pdn0001-
vol1.pdf. Accessed July 3, 2009.
32. Bioavailability and Bioequivalence. Title 21 Code of Federal Regulations. Part 320.
Washington, DC: U.S. Government Printing Office. Revised as of April 1, 2008.
33. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Guidance for Industry: Nevirapine. May
2008. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatory
Information/Guidances/ucm088752.pdf. Accessed June 21, 2009.
34. Chen, ML, Shah V, Patnaik R, et al. Bioavailability and bioequivalence: An FDA regu-
latory overview. Pharm Res 2001; 18:1645–1650.
35. Blume H, McGilveray IJ, Midha KK. BIO-International ‘94 Conference on Bioavail-
ability, Bioequivalence and Pharmacokinetic Studies and Pre-Conference Satellite on
“In Vivo/In Vitro Correlation.” Munich Germany, June 14–17, 1994. Eur J Drug Metab
Pharmacokinet 1995; 2:3–13.
36. El-Tahtawy AA, Jackson AJ, Ludden TM. Evaluation of bioequivalence of highly-
variable drugs using Monte Carlo simulations. I. Estimation of rate of absorption for
single and multiple-dose trials using Cmax. Pharm Res 1995; 12:1634–1641.
37. El-Tahtawy AA, Jackson AJ, Ludden TM. Comparison of single and multiple dose
pharmacokinetics using clinical bioequivalence data and Monte Carlo simulations.
Pharm Res 1996; 11:1330–1336.
38. El-Tahtawy AA, Tozer TN, Harrison F, et al. Evaluation of bioequivalence of highly
variable drugs using clinical trial simulations. II. Comparison of single- and multiple-
dose trials using AUC and Cmax. Pharm Res 1998; 15:98–104.
39. Wagner JG. Nonlinear pharmacokinetics: Examples of anticancer agents and implica-
tions in bioavailability. In: Sharma KN, Sharma KK, Sen P, eds. Generic Drugs, Bioe-
quivalence and Pharmacokinetics. New Delhi, India: University College of Medical
Sciences, 1990:111–118.
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
40. US Dept of Health and Human Services, Food and Drug Administration, Center for
Drug Evaluation and Research. Guidance for Industry: Phenytoin Sodium, May 2008.
http://www.fda.gov/cder/guidance/bioequivalence/default.htm. Accessed July 3,
2009.
41. Clinical Pharmacology Biopharmaceutics Review(s), September 28, 2000. Mifepri-
stone. Drugs@FDA. http://www.accessdata.fda.gov/drugsatfda docs/nda/2000/
20687 mifepristone.cfm. Accessed July 3, 2009.
42. U S Dept of Health and Human Services, Food and Drug Administration, Center
for Drug Evaluation and Research. Guidance for Industry: Food-Effect Bioavail-
ability and Fed Bioequivalence Studies. December 2002. http://www.fda.gov/
downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/
ucm070241.pdf. Accessed July 3, 2009.
43. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Draft Guidance for Industry: Risedronate
Sodium. December 2008. http://www.fda.gov/downloads/Drugs/Guidance
ComplianceRegulatoryInformation/Guidances/ucm089588.pdf. Accessed July 3,
2009.
44. Davit BM, Conner DP. Food Effects on Drug Bioavailability: Implications for New and
Generic Drug Development. In: Krishna R, Yu L, eds. Biopharmaceutics Applications
in Drug Development. New York, NY: Springer, 2008:317–335.
45. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Guidance for Industry: Efavirenz. May
2008. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatory
Information/Guidances/ucm086238.pdf. Accessed July 3, 2009.
46. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Guidance for Industry: Didanosine. May
2008. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatory
Information/Guidances/ucm085602.pdf. Accessed July 3, 2009.
47. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Guidance for Industry: Mefloquine. May
2008. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatory
Information/Guidances/ucm088650.pdf. Accessed July 3, 2009.
48. US Dept of Health and Human Services, Food and Drug Administration,
Center for Drug Evaluation and Research. Draft Guidance for Industry: Ima-
tinib Mesylate. March 2009. http://www.fda.gov/downloads/Drugs/Guidance
ComplianceRegulatoryInformation/Guidances/ucm118261.pdf. Accessed June 22,
2009.
49. US Dept of Health and Human Services, Food and Drug Administration,
Center for Drug Evaluation and Research. Draft Guidance for Industry:
Methotrexate Sodium. April 2009. http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/UCM148209.pdf.
Accessed June 22, 2009.
50. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Draft Guidance for Industry: Alen-
dronate Sodium. January 2008. http://www.fda.gov/downloads/Drugs/Guidance
ComplianceRegulatoryInformation/Guidances/ucm082421.pdf. Accessed July 3,
2009.
51. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Guidance for Industry: Potassium Chlo-
ride Modified-Release Tablets and Capsules – In Vivo Bioequivalence and In
Vitro Dissolution Testing. October, 2005. http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/ucm072890.pdf. Accessed
July 3, 2009.
52. US Dept of Health and Human Services, Food and Drug Administration, Center
for Drug Evaluation and Research. Guidance for Industry: Topical Dermato-
logic Corticosteroids: In Vivo Bioequivalence. June 1995. http://www.fda.gov/
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm
070234.pdf. Accessed July 3, 2009.
53. FDA Center for Drug Evaluation and Research – Final Response Letter,
Docket No. FDA-2007-P-0418. May 7, 2008. http://www.regulations.gov/
fdmspublic/component/main?main = DocumentDetail&o = 09000064805529eb.
Accessed July 3, 2009.
54. FDA Center for Drug Evaluation and Research – Final Response Let-
ter, Dockets No. 2004P-206/CP1; 2004–239/CP1, SUP 1, SUP 2 & PSA 1;
2004P-0348/CP1 & SUP 1; and 2004P-0523/CP1 & PSA1. February 22, 2006.
http://www.regulations.gov/fdmspublic/component/main?main = DocumentDe-
tail&o = 090000648046ffd5. Accessed July 3, 2009.
55. US Dept of Health and Human Services, Food and Drug Administration,
Center for Drug Evaluation and Research. Interim Guidance for Industry:
Cholestyramine Powder In Vitro Bioequivalence. July 1993. http://www.fda.gov/
downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/
ucm070230.pdf. Accessed June 22, 2009.
56. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Draft Guidance for Industry: Seve-
lamer Hydrochloride. July 2008. http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/ucm089621.pdf. Accessed
July 3, 2009.
57. Swenson CN, Fundak G. Observational cohort study of switching warfarin sodium
products in a managed care organization. Am J Health Syst Pharm 2000; 57:
452–455.
58. 57 Fed Regist 29354, July 1, 1992.
59. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Draft Guidance for Industry: Arip-
iprazole. November 2007. http://www.fda.gov/downloads/Drugs/Guidance
ComplianceRegulatoryInformation/Guidances/ucm082554.pdf. Accessed July 3,
2009.
60. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Guidance for Industry: Lamotrigine. May
2008. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatory
Information/Guidances/ucm086283.pdf. Accessed June 22, 2009.
61. US Dept of Health and Human Services, Food and Drug Administration, Cen-
ter for Drug Evaluation and Research. Guidance for Industry: Waiver of In Vivo
Bioavailability and Bioequivalence Studies for Immediate-Release Solid Oral Dosage
Forms Based on a Biopharmaceutics Classification System. August 2000. http://
www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/
Guidances/ucm070246.pdf. Accessed July 3, 2009.
62. Committee Discussion. FDA Advisory Committee for Pharmaceutical Science
Meeting Transcript. April 13–14, 2004. http://www.fda.gov/ohrms/dockets/ac/
04/minutes/4034M1.htm. Accessed July 3, 2009.
63. Benet L. Therapeutic Considerations of Highly Variable Drugs. FDA Advisory
Committee for Pharmaceutical Science Meeting Transcript. October 6, 2006.
http://www.fda.gov/ohrms/dockets/ac/06/slides/2006–4241s2-index.htm.
Accessed July 3, 2009.
64. Haidar SH, Davit B, Chen ML, et al. Bioequivalence approaches for highly variable
drugs and drug products. Pharm Res 2008; 25:237–241.
65. Haidar SH, Makhlouf F, Schuirmann DJ, et al. Evaluation of a scaling approach for
the bioequivalence of highly variable drugs. AAPS J 2008; 10:450–454.
66. Davit B. Outliers and Inadequate Profiles in Bioequivalence Studies – US FDA
Perspective. Health Canada Scientific Advisory Committee on Bioavailability and
Bioequivalence Record of Proceedings. June 3–4, 2004. http://www.hc-sc.gc.ca/dhp-
mps/alt formats/hpfb-dgpsa/pdf/prodpharma/sacbb rop ccsbb crd 2004–06-03-
eng.pdf. Accessed July 3, 2009.
SPH SPH
IHBK055-12 IHBK055-Kanfer January 22, 2010 9:18 Char Count=
Henrike Potthast
Federal Institute for Drugs and Medical Devices, Bonn, Germany
BACKGROUND
Much progress has been achieved over the last 50 years in the field of phar-
maceuticals, both in terms of introducing new medicines and improving the
regulation of medicines. This progress involves mostly highly industrialized
countries where citizens can benefit from new innovative drugs and enjoy
access to quality assured multisource (generic) medicines as well. Lack of access
to quality essential drugs, the majority of which are multisource (generic)
medicines, remains a serious health problem and global disequilibrium of
quality continues to threaten patients in many parts of the world (1). The overall
tendency is that resource-constrained or resource-poor countries are less likely
to control the quality of products on the market, enjoy political support for the
regulators, or have properly resourced and functioning regulatory authorities
(2). It is no wonder that in many resource-poor settings patients do not trust
locally authorized multisource (generic) products.
In terms of what is required for regulatory approval of medicines it is
important to distinguish between two major groups, innovative new medicines
(new chemical entities or NCEs) and multisource (generic) medicines. To launch
an innovator product the manufacturer/applicant has to pass rigorous scientific
assessment by the competent regulatory authorities and prove its product’s qual-
ity, safety, and efficacy. The most difficult aspect of these is to prove the safety
and efficacy of the new drug since that has to be based on original preclinical
a The WHO is the directing and coordinating authority for health within the United Nations
system. It is responsible for providing leadership on global health matters, shaping the health
research agenda, setting norms and standards, articulating evidence-based policy options, pro-
viding technical support to countries and monitoring and assessing health trends.
b The views stated in this chapter reflect the views of the authors and not necessarily those of
the WHO.
282
SPH SPH
IHBK055-13 IHBK055-Kanfer January 6, 2010 20:9 Char Count=
at its site of action but, since it is generally agreed that the API in the systemic
circulation is usually in some kind of equilibrium with its site of action, sys-
temic concentrations may serve to evaluate bioavailability. Accordingly, concen-
trations derived after intravenous administrations define the 100% standard, that
is, “absolute” bioavailability. Therefore, comparing systemic concentrations after
the administration of an oral dosage form to those obtained following intra-
venous administration results in “absolute bioavailability” of the oral product.
However, it is not possible to distinguish between formulation properties and its
impact on the pharmacokinetics of the API by means of investigating absolute
bioavailability. In contrast relative bioavailability is meant to be the comparison
of dosage forms other than intravenous and one of the most interesting is com-
paring an aqueous oral solution with a solid oral dosage form. The result of such
a comparison may serve to describe the formulation impact of the solid dosage
form, for example, possibly formulation-related diminished availability. Hence,
comparing bioavailability becomes basic for the assessment of bioequivalence of
systemically acting (particularly orally administered) generics [i.e., multisource
pharmaceutical drug products (MPPs)]. According to the latest WHO guidance
on the topic, Annex 7 of the Technical Series Report No. 937 (5) the term bioe-
quivalence is defined as similar bioavailabilities of pharmaceutical equivalent or
pharmaceutical alternative products in terms of peak (Cmax and Tmax ) and total
exposure (AUC) after administration of the same molar dose under the same
conditions to such a degree, that their effects can be expected to be essentially the
same. In other words, essentially similar products should be interchangeable.
product, but it is necessary that it be treated as a new product [see Section 6.11.2
in Ref. (5)].
Another difficulty may arise in cases when no innovator/originator prod-
uct is available for some reason. The appropriate selection of the comparator as
recommended by WHO is discussed in “Choice of Comparator Products” section
(vide infra).
For completeness it should be noted that bioequivalence testing is required
not only for new MPP applications but also in the case of major formulation
changes (variations). For modified MPPs, bioequivalence has to be demonstrated
by comparison against the designated reference comparator product. A study
comparing the modified MPP to the originally approved (unchanged) MPP is
not sufficient as this could lead to “creep” away from the connection between the
reference product and the MPP, the link upon which the evidence for the safety
and efficacy of the MPP is based. The latter comparison is an adequate option
only for innovator products (see “Modified-Release MPPs” section [vide infra]).
Formulation-Related Biowaiver
Section 4 of the WHO Technical Report of 2006, Annex 7 (5) outlines those cases
when bioequivalence studies are not necessary due to the particular formulation
and/or site of administration and/or intended effect. Accordingly, aqueous solu-
tions intended for oral or intravenous/parenteral administration are exempted
from bioequivalence testing since no formulation effect is expected even in the
case when slightly different excipients have been used (i.e., buffer, preserva-
tive, antioxidant). Other soluble formulations (e.g., syrups, tinctures, powders
for reconstitution) except suspensions may be treated likewise.
Exemption of bioequivalence testing is also generally acceptable for phar-
maceutically equivalent products such as gases, otic or ophthalmic formula-
tions, and topical products if they are not intended to be systemically effective,
aqueous nebulizer inhalation products or aqueous-based nasal sprays. Hence,
formulation-related biowaivers may be applicable if pharmaceutical product
quality is deemed sufficient to assure therapeutic equivalence when no formu-
lation effect is expected. Notwithstanding, however, the investigative products
should contain the same API in the same molar concentration, whereas any
possible impact of difference in excipients used will have to be appropriately
addressed by the applicant (Table 2).
can be made as a part of the authorization process for MPPs, WHO has published
a guidance that includes a list of comparator products derived from information
collected from drug regulatory authorities and the pharmaceutical industry (6).
These “International Comparator Products” or WHO comparator products can
be selected by national authorities when a “nationally authorized innovator” is
not available. This guidance also suggests criteria, in a decision-tree format, that
can be used in the selection of a comparator product.
IN VIVO APPROACHES
As discussed earlier, in vivo documentation of bioequivalence is especially
important for certain medicines and dosage forms, and the pharmacokinetic
bioequivalence study is considered to be the most effective and sensitive design
for achieving this purpose.
There are many issues to be considered when designing a bioequivalence
study to compare the in vivo performance of two products, however, the ultimate
goal is to design a study that minimizes the variability that is not attributable to
the formulations being compared and to eliminate any bias in the study. In gen-
eral, studies comparing product performance following a single administration
of each product, that is, single-dose bioequivalence studies, are considered to
be the most effective study designs for the purpose of comparing a MPP to a
comparator product.
SPH SPH
IHBK055-13 IHBK055-Kanfer January 6, 2010 20:9 Char Count=
Comparator Product
The product to which the MPP will be compared in a bioequivalence study
should be determined as discussed in Choice of Comparator Products section.
Study Design
Cross-Over Versus Parallel Designs
Although there are several options to be considered, the study design of choice
for comparing a MPP to a comparator product should generally be a random-
ized, two-period, two-sequence, single-dose, cross-over bioequivalence study
conducted in healthy subjects under fasted conditions. Given that greater vari-
ability is generally observed in pharmacokinetic comparisons made between
subjects than those made repeatedly within a subject, a cross-over study design
is recommended to take advantage of the lower variability associated with intra-
subject comparisons. That is, using an intra-subject comparison to study product
performance will help minimize the variability in the data that is attributable to
factors other than the products themselves. It is recommended that the washout
period between doses in a cross-over design be at least five times the terminal
half-life of the active ingredient.
Food Considerations
As mentioned earlier, one of the primary considerations in the design of a bioe-
quivalence study is minimizing the variability in the data that is attributable to
factors other than the products themselves. The presence of food in the GI tract at
the time of drug administration can introduce considerable variability into phar-
macokinetic data because of the multiple and complicated effects food can have
on both the physiology of the GI tract, the disintegration of the drug product, and
the dissolution of the API. However, although a study conducted under fasted
conditions is preferred, there are circumstances under which a study conducted
under fed conditions may be accepted. For example, if the active ingredient is
known to cause significant GI disturbance when administered under fasted con-
ditions, or if the product labelling clearly restricts administration to the fed state,
then a study conducted under fed conditions should be used to assess bioequiv-
alence. In such fed studies, the test meal selected should be designed to account
for local custom and diet, should be consumed by subjects within a 20-minute
time frame, and drug administration should follow within 30 minutes.
The bioequivalence comparison of a proposed modified-release (MR) for-
mulation is discussed in Modified-Release MPPs section but, briefly with respect
to food, the comparison of a MR formulation to the appropriate comparator
product must be investigated under both fasted and fed conditions. In such situ-
ations, the test meal employed in the fed study should be designed to challenge
the robustness of the proposed MR formulation by promoting a maximal pertur-
bation of the GI conditions relative to the fasted state, for example, a high-fat,
high-calorie meal.
Participants
(a) Number of Subjects: The number of subjects required for a successful bioe-
quivalence study should be determined based on the standards that must
be met (see Section Handling of Study Data below) and the drug products
being compared. The probability that a study of a given size will meet the
applicable standards will depend on the expected mean difference between
the test and reference formulations and variability associated with the drug
involved, that is, the anticipated intrasubject coefficient of variation for a
cross-over design.
A justification for the number of subjects enrolled in a study, which
includes a sample size calculation, should always be provided in the study
protocol. A minimum of 12 subjects is required for all studies.
(b) Subject Selection: Bioequivalence studies should normally employ healthy
volunteers. The volunteers should be standardized based on characteristics
SPH SPH
IHBK055-13 IHBK055-Kanfer January 6, 2010 20:9 Char Count=
such as age, height, and weight. If the product under development is pro-
posed for use in both sexes, it is suggested that both male and female vol-
unteers be recruited for the study.
In situations where administration of the study drug to healthy volun-
teers is not acceptable due to its potency or toxicity, a study employing a
patient population may be necessary. Such studies will usually involve a
multiple-dose study design, as discussed below.
Bioanalytical Methods
The validity of the study conclusions depends on the reliability and reproducibil-
ity of the data collected. Therefore, all analytical methods used to measure the
active ingredient in the chosen biological fluid must be well characterized, fully
validated, and documented.
Bioanalytical methods must meet the requirements of specificity, sensi-
tivity, accuracy, precision, and reproducibility. Investigators are encouraged to
adhere to the recommendations of the Bioanalytical Method Validation Confer-
ence (10) with respect to both prestudy method validation and within-study clin-
ical sample analyses and accompanying quality control.
The analytical method, validation procedures, and acceptance/rejection
criteria must be clearly defined in the analytical protocol and associated stan-
dard operating procedures (SOPs) prior to the conduct of a study.
A dossier should include a complete Method Validation Report, copies
of all applicable SOPs, and a complete analytical report. The analytical report
should summarize the results of the analysis of the clinical study samples along
with complete details of the calibration and quality control sample analyses,
repeat analyses as per SOP, and a representative sample of chromatograms from
the study.
Monitoring of Metabolites
The purpose of a bioequivalence study is to compare the performance of two or
more products. The most sensitive approach to achieving this goal is to mon-
itor the parent compound’s disposition in the systemic circulation, that is, the
API being released from the products. Therefore, the API is the analyte of choice
for a bioequivalence study. If, employing up-to-date analytical methodologies,
the API cannot be measured accurately in the biological matrix over a suffi-
cient period of time to properly characterize a concentration–time profile, mea-
surement of the primary, therapeutically active metabolite may be justified. The
choice of analyte must be established a priori and stated in the study protocol.
Should it be found appropriate to monitor the metabolite, the study design, for
example, washout period, should be adjusted appropriately.
Acceptance Ranges
(a) Area under the curve (AUC): The 90% confidence interval for the relative
mean AUCT of the test to the reference product should be within 0.80 to
1.25. If the therapeutic range is particularly narrow, the acceptance range
may need to be reduced based on a clinical justification. A larger acceptance
range may be acceptable in exceptional cases if justified clinically.
(b) Maximal Concentration (Cmax ): The 90% confidence interval for the rela-
tive mean Cmax of the test to the reference product should be within 0.80 to
1.25. As the measurement of Cmax is inherently more variable than the mea-
surement of AUC, Annex 7 (5) suggests that there are certain cases where
a wider acceptance range may be justified, for example, see “Highly Vari-
able Drugs” section later in the chapter. The range used must be defined
a priori and should be justified, taking into account safety and efficacy
considerations.
(c) Time to Maximal Concentration (Tmax ): The WHO guidances indicate that
the statistical evaluation of Tmax is necessary only if there is a clinically rele-
vant claim with respect to time of onset of action or concerns about adverse
events. In such a case, the nonparametric 90% confidence interval for the
relative Tmax measure should lie within a clinically relevant range.
established that the drug in question is a highly variable drug and that there are
no concentration-related concerns with regard to the safety or efficacy profiles
of the drug. A widening of the acceptance range for AUC may be considered in
exceptional circumstances if justified clinically.
Outliers
Any statistical methods to be used for the detection of outlier data must be clearly
defined in the study protocol. The statistical recognition of data as an outlier is
not considered to be a justification for its exclusion but, an indication that a fur-
ther examination of these data is appropriate. The criteria that can be employed
to justify exclusion should be defined a priori and, if an additional examination
is undertaken to investigate outlying data, this examination must be extended to
all subjects to ensure the uniform treatment of the data.
Within-clinical setting issues, for example, significant protocol violations,
and/or physiological/medical explanations for the aberrant data, should be
sought and would be a critical element in a justification for excluding outlier data
from the main dataset. Should exclusion of the outlier data be proposed, com-
plete statistical analyses with and without the outlier data should be included as
an appendix to the study report.
Multiple-Dose Studies
Should it be found necessary to perform a steady-state, multiple-dose study,
the basic study design considerations discussed earlier would be applicable
for the dosing period during which blood samples are collected. Annex 7 (5)
describes situations where a multiple-dose study may be appropriate, for exam-
ple, a multiple-dose study in patients may be required when the API is too potent
or too toxic to be administered in healthy volunteers. Other situations in which
multiple-dose studies might be appropriate include the following:
r Drugs for which assay sensitivity is too low to adequately characterize the
concentration–time disposition profile after a single dose
r Products involving drugs that exhibit nonlinear kinetics at steady state
r MR products with a tendency to accumulate. In the latter case, the multiple-
dose study would be required in addition to single-dose studies.
period following the last dose of the first product can overlap with the approach
to steady state of the second product provided this period between drug admin-
istrations is sufficiently long, that is, at least three times the terminal half-life of
the API. Blood samples should be collected to establish that steady state has been
achieved.
With regard to metrics for the assessment of bioequivalence, the parame-
ters Cmax , Cmin (the minimum concentration observed in the dosing interval), the
peak-trough fluctuation (the percentage difference between Cmax and Cmin ) and
the AUC of the dosing interval being sampled (AUC ) should be calculated. The
calculated Cmax and AUC parameters should meet the standards discussed ear-
lier, while the relative mean Cmin at steady state of the test to reference product
should not be less than 0.80.
Immediate-Release MPPs
As discussed above, a single-dose study under fasting conditions is generally
regarded as the most conservative and sensitive approach to assess the rate and
extent of absorption, that is, comparative bioavailability between a test and ref-
erence product. However, when the drug product should not be taken on an
empty stomach and the innovator’s SPC indicates so, it may be appropriate to
do the comparison under standardized fed conditions. Similarly in the case when
there are tolerability concerns under fasting conditions, the study may need to be
conducted under fed conditions. In general, for immediate-release MPPs steady-
state studies are only acceptable in rare cases, that is, they should be performed in
addition to single dose studies if necessary and/or possible (see “Multiple-Dose
Studies” section).
It may be possible to waive the in vivo comparison if the MPP con-
tains a drug substance that has been classified as being eligible in terms
of possible therapeutic risks, solubility and permeability according to the
SPH SPH
IHBK055-13 IHBK055-Kanfer January 6, 2010 20:9 Char Count=
Modified-Release MPPs
To evaluate the rate and extent of absorption in a most sensitive experimental
setting, single dose studies are also relevant to assess bioequivalence for MR
products. Multiple-dose studies are sometimes required in addition, particularly
if accumulation after multiple dosing is expected or if fluctuation is considered
important. In contrast to the considerations for immediate-release formulations,
the investigation of food effects is relevant for MR products as it is critical to test
the formulation-related performance of the products under fasting and fed con-
ditions, that is, to test both ends of the spectrum of conditions under which the
product may be used once approved. Testing under the range of possible GI con-
ditions is necessary to monitor for significant changes in product performance
under the varying conditions, in particular to exclude the risk of dose dumping,
that is, the unintended immediate drug release from a MR dosage form, due to
the influence of concomitant food intake. Therefore, in addition to fasting stud-
ies, pharmacokinetic bioequivalence studies conducted under fed conditions are
usually required for MR MPPs.
It should be noted that a BCS-based biowaiver is generally not acceptable
for modified-release formulations.
Comparative pharmacodynamic studies or clinical trials are alternatives
when pharmacokinetic bioequivalence studies cannot be performed, for exam-
ple, for safety and/or tolerability reasons.
BCS-Based Biowaiver
The possibility of “in vitro documentation of bioequivalence” for “certain
medicines and dosage forms” is specified in Section 9 of the WHO guidance
document (5). If the drug substance in question is highly soluble and highly per-
meable (BCS class I) and is manufactured as an immediate-release dosage form,
exemption from an in vivo pharmacokinetic bioequivalence study may be con-
sidered provided that relevant dissolution requirements are fulfilled.
The in vitro approach basically refers to the BCS. Accordingly, active phar-
maceutical ingredients are classified into classes based on their aqueous solubil-
ity and permeability characteristics (Table 3).
It should be noted that solubility is not meant to be the absolute solubility
here. In contrast, high solubility refers to the highest single unit dose to be com-
pletely soluble in 250 mL aqueous buffer medium within the pH range of 1.2 to
6.8 without any stability problems.
As another related physicochemical characteristic, high permeability
should be demonstrated for the particular API demonstrating that the fraction
of the dose absorbed amounts to at least 85%. Accordingly, high permeability
would stand for almost complete absorption of the compound in humans.
SPH SPH
IHBK055-13 IHBK055-Kanfer January 6, 2010 20:9 Char Count=
Proportionality-Based Biowaivers
Another approach to waive in vivo bioequivalence testing may be taken when
an MPP is intended for marketing in different strengths manufactured in pro-
portionally formulated dosage forms as opposed to an application for a single
strength only. It should be noted that dose proportionality is defined in two
ways according to Section 9.3.1 of the WHO guidance (5). Either exact propor-
tional composition of excipients is evident or different strengths are obtained by
altering only the amount of the API which is solely adequate for high potency
drugs. The latter case applies if the amount of API is relatively low (<10 mg per
dosage unit), the total weight of the dosage form remains nearly the same for
all strengths of the product series, the same excipients are used for all strengths
and the change in strength is obtained by altering the amount of API only (5).
Biowaivers based on dose proportionality may be considered if one in vivo bioe-
quivalence study was performed, usually on the highest dose strength of the
product series in question. Convincing comparative in vitro dissolution data are
required to link the tested biobatch of the MPP, which was proven bioequivalent
to the comparator and the additional proportional strengths.
Proportionality-based biowaivers differ from BCS-based biowaivers in that
the former approach is eligible (in principle) for immediate- and extended-
release formulations whilst the latter can be considered for specific drug sub-
stances in immediate-release oral dosage forms only.
this chapter, it should be mentioned that iviv correlations are always product
related, that is, they cannot replace bioequivalence investigations between dif-
ferent products (e.g., MPP vs. comparator) from different applicants. In contrast,
an iviv correlation established for a particular product may help to set meaning-
ful specifications and also obviate the need for bioequivalence studies for certain
postapproval changes.
CONCLUSIONS
The basic concept of bioequivalence testing has been implemented in interna-
tional guidelines for decades although requirements have become more detailed
and specific over time. In vitro approaches are nowadays considered to further
facilitate production of affordable medicines worldwide. However, meaningful
bioequivalence testing requires thorough planning and a decision regarding the
approach which may be applicable considering the specific characteristics of a
particular drug substance and/or drug product. Development of science, regu-
latory practice, and therapeutic experience will guide the way forward.
REFERENCES
1. Rägo L. Global disequilibrium of quality. In: Prince R, ed. Pharmaceutical Quality.
Illinois: Davies Health Care International Publishing, 2004:3–21.
2. Ratanwijitrasin S, Wondemagegnehu E. Effective Drug Regulation. A multicountry
Study. Geneva, Switzerland: World Health Organization, 2002:1–142.
3. Kopp S, Rägo L. The International Pharmacopoeia in the changing environment.
Pharm Policy and Law 2007; 9:357–368.
4. Ritschel WA, Kearns GL. Handbook of Basic Pharmacokinetics — Including Clinical
Applications, 5th ed. Washington, DC: American Pharmaceutical Association, 1999.
5. Multisource (generic) pharmaceutical products: Guidelines on registration require-
ments to establish interchangeability. In: WHO Expert Committee on Specifications
for Pharmaceutical Preparations, Fortieth Report. Geneva, Switherland: World Health
Organization, 2006 (WHO Technical Report Series, No. 937, Annex 7):347–390.
6. Guidance on the selection of comparator pharmaceutical products for equivalence
assessment of interchangeable multisource (generic) products. In: WHO Expert
Committee on Specifications for Pharmaceutical Preparations, Thirty-sixth Report.
Geneva, Switzerland: World Health Organization, 2002 (WHO Technical Report
Series, No. 902):161–180.
7. van Faassen F, Vromans H, et al. Biowaivers for oral immediate-release products:
Implications of linear pharmacokinetics. Clin Pharmacokinet 2004; 43:1117.
8. Additional guidance for organizations performing in vivo bioequivalence studies.
In: WHO Expert Committee on Specifications for Pharmaceutical Preparations, Forti-
eth Report. Geneva, Switzerland: World Health Organization, 2006 (WHO Technical
Report Series, No. 937, Annex 9):439–461.
9. Information for Applicants on the Choice of Comparator Products for the Prequali-
fication Project. Located on the World Health Organization (WHO) Prequalification
of Medicines Web site, Guidance on selection of comparator product page. http://
apps.who.int/prequal/info applicants/info for applicants BE comparator.htm
10. Shah VP et al. Bioanalytical method validation—A revisit with a decade of progress.
Pharm Res 2000; 17:1551–1557.
11. Schuirmann DJ. A comparison of the two one-sided tests procedure and the power
approach for assessing the equivalence of average bioavailability. J Pharmacokinet
Biopharm 1987; 15:657–680.
12. Westlake WJ. Bioavailability and bioequivalence of pharmaceutical formulations. In:
Peace KE ed. Biopharmaceutical Statistics for Drug Development. New York: Marcel
Dekker, Inc., 1988:329–352.
SPH SPH
IHBK055-13 IHBK055-Kanfer January 6, 2010 20:9 Char Count=
13. Guidelines for registration of fixed-dose combination medicinal products. In: WHO
Expert Committee on Specifications for Pharmaceutical Preparations, Thirty-ninth
report. Geneva, World Health Organization, 2005 (WHO Technical Report Series, No.
929, Annex 5):94–142.
14. Proposal to waive in vivo bioequivalence requirements for WHO Model List of Essen-
tial Medicines immediate-release, sold oral dosage forms. In: WHO Expert Commit-
tee on Specifications for Pharmaceutical Preparations, Fortieth Report. Geneva, World
Health Organization, 2006 (WHO Technical Report Series, No. 937, Annex 8):391–439.
15. Stosik AG, Junginger HE, Kopp S, et al. Biowaiver monographs for immediate release
solid oral dosage forms: Metoclopramide hydrochloride [published online ahead of
print February 12, 2008]. J Pharm Sci.
16. Becker C, Dressman JB, Amidon GL, et al. Biowaiver monographs for immediate
release solid oral dosage forms: Pyrazinamide [published online ahead of print Febru-
ary 12, 2008]. J Pharm Sci.
17. Becker C, Dressman JB, Amidon GL, et al. Biowaiver monographs for immediate
release solid oral dosage forms: Ethambutol dihydrochloride [published online ahead
of print August 21, 2007]. J Pharm Sci.
18. Becker C, Dressman JB, Amidon GL, et al. Biowaiver monographs for immediate
release solid oral dosage forms: Isoniazid. J Pharm Sci 2007; 96:522–531.
19. Emami J. In vitro-in vivo correlations: From theory to applications. J Pharm Pharma-
ceut Sci 2006; 9:169.
SPH SPH
IHBK055-IND IHBK055-Kanfer January 21, 2010 9:56 Char Count=
Index
301
SPH SPH
IHBK055-IND IHBK055-Kanfer January 21, 2010 9:56 Char Count=
302 Index
Index 303
304 Index
Enantiomers versus racemates, 190 Generic drug review process, in Taiwan, 234
Ethics Committee, 22, 156, 157 bioequivalence review process, 236
Ethics Committee on Assisted Reproductive labeling review process, 235
Technology, 22 quality review process, 235, 236
European Medicines Agency (EMEA), 96, Generic medicines, 116–118
106 Generic substitution, 9–11
European Union (EU), 3, 4, 95, 97 Genetic phenotyping, 248
fixed combination drug products, 108, Global harmonization, 109, 110
109 Global Malaria Program, 284
global harmonization, 109, 110 Good clinical practice (GCP), 212, 286
modified release oral and transdermal Good laboratory practice (GLP), 140
dosage forms, 107 Good manufacturing practice (GMP), 19, 51,
delayed release oral formulations, 108 165, 211, 212, 232, 233
prolonged release oral formulations, 107, Group sequential designs/add-on studies, of
108 WHO, 294
transdermal drug delivery systems, 108 Guinea pig maximization test (GPMT), 146
oral immediate release dosage forms with
systemic action, 97, 98 Hatch–Waxman Amendments (1984), 258, 259
BE metrics, 101–104 Health Canada guidance, 71, 76, 79
exemptions from in vivo BE studies, Health Canada policy, 88
104–106 Health Research Council Ethics Committee
locally applied drug products, 106, 107 (HRCEC), 22
study design, 98–101 Highly variable drugs (HVDs), 34, 271, 272,
Expert Advisory Committee (EAC), 67, 68 293
Expert Advisory Committee on HPFB of Canada, 103
Bioavailability (EAB), 68 HRC Guidelines for Ethics Committee
Accreditation, 23
Failed bioequivalence studies, 272–275 Human Research Ethics Committee (HREC),
Fed bioequivalence studies, 263, 264 23
Fed/fasting studies Human skin blanching assay (HSBA), 40, 73,
in India, 123 85
in South Africa, 185, 186
FIFARMA (Federación Latinoamericana de la In vitro cytogenetic assay, 147, 148
Industria Farmacéutica), 212 In vitro–in vivo correlations, 298, 299
Fixed combination drug products, 108, 109 In vitro studies, in India, 133, 134
Fixed dose combinations (FDC), 116, 117, 198, In vivo bioequivalence studies, waivers of,
289, 295 180
Fluticasone propionate, 268 controlled/modified release dosage forms,
Food and Drug Administration (FDA), 6–8, 64
95, 254–256, 258–260264, 267, 270, 272, immediate release drug products, 64
276 In vivo cytogenetic assay, 148
Foreign bioequivalence data, acceptance of, In vivo micronucleus assay, 148
180 Immediate release drug products, 269
Foreign reference products, 193, 204 Biopharmaceutics Classification System
Formulation performance, 255 (BCS), 89
Formulation-related biowaiver, 287 controlled/modified-release dosage forms,
89, 90
General Directorate of Drug and Pharmacy different strength dosage forms, 88, 89
(GDDP), 242 Immediate-release MPPs, 295, 296
Generic Drug Advisory Committee, 260, 261 Immediate-release products, 198, 202
Generic drug products, 4, 211, 243 Import license application, 118
SPH SPH
IHBK055-IND IHBK055-Kanfer January 21, 2010 9:56 Char Count=
Index 305
306 Index
Index 307
308 Index
Index 309